CA3204457A1 - Mdma in the treatment of alcohol use disorder - Google Patents

Mdma in the treatment of alcohol use disorder Download PDF

Info

Publication number
CA3204457A1
CA3204457A1 CA3204457A CA3204457A CA3204457A1 CA 3204457 A1 CA3204457 A1 CA 3204457A1 CA 3204457 A CA3204457 A CA 3204457A CA 3204457 A CA3204457 A CA 3204457A CA 3204457 A1 CA3204457 A1 CA 3204457A1
Authority
CA
Canada
Prior art keywords
mdma
subject
psychotherapy
alcohol
assisted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CA3204457A
Other languages
French (fr)
Inventor
Ben SESSA
Laurie HIGBED
David Nutt
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Awakn Ls Europe Holdings Ltd
Original Assignee
Awakn Ls Europe Holdings Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Awakn Ls Europe Holdings Ltd filed Critical Awakn Ls Europe Holdings Ltd
Publication of CA3204457A1 publication Critical patent/CA3204457A1/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/357Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having two or more oxygen atoms in the same ring, e.g. crown ethers, guanadrel
    • A61K31/36Compounds containing methylenedioxyphenyl groups, e.g. sesamin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse

Abstract

The invention discloses compositions of MDMA and methods of their use in MDMA-assisted psychotherapy for the treatment of patients with alcohol use disorder (AUD) including physically dependent alcohol use disorder ("dependent use" or "alcohol addiction") and non-physically dependent alcohol use disorder ("harmful use").

Description

MDMA IN THE TREATMENT OF ALCOHOL USE DISORDER
INVENTOR: Ben Sessa APPLICANT: Awakn Life Sciences Inc.
DECLARATION OF PRIORITY
[01] This application claims priority under PCT Article 8(1) and PCT Rule 4.10 to U.S. Provisional Application No. 63/134,582, filed January 6, 2021, and U.S.
Provisional Application No. 63/137,615, filed January 14, 2021, both of which are incorporated by reference for all purposes as if fully set forth herein.
TECHNICAL FIELD
[02] Described herein are compositions of MDMA and methods of their use in MDMA-assisted psychotherapy for the treatment of patients with alcohol use disorder (AUD) including physically dependent alcohol use disorder ("dependent use" or "alcohol addiction") and non-physically dependent alcohol use disorder ("harmful use").
BACKGROUND OF THE INVENTION
[03] The cost of excessive alcohol use is high, both to the individual, in terms of physical and mental health, and to society in terms of social and medical care.
Evidence indicates that this is a growing problem with almost a quarter of adults in the ITK now labelled as problem drinkers, and similar statistics elsewhere worldwide. Despite a wide variety of psychological and pharmacological treatment options, long term outcomes are poor, with high rates (up to 60% at 12 months and 80% at 3 years) of relapse after medical detoxification and dedicated treatment programs (Evren et al. 2010, Moos & Moos 2006). And in the context of the current COVID-19 pandemic, attention to the issue of best management of AUD has become even more pertinent, as increasing evidence is emerging that people's normal drinking levels are rising steeply (Cargill 2020; Clay & Parker 2020). At the time of this filing, deaths from alcohol in the UK were up almost 19%, the largest single-year increase recorded (ONS
2021).
[04] The impact of alcohol misuse is widespread, encompassing alcohol related illness and injuries as well as significant social impacts to family, friends and the wider community. People
5 with AUD broadly commonly present with high levels of depression, social anxiety and social exclusion (Castillo-Camiglia et al. 2019). Additionally, in patients with physical dependence, studies indicate that repeated detoxifications are associated with more complications (Malcolm et al. 2000). This highlights further the need for successful treatments to help patients remain abstinent after their first detoxification and avoid the complications that can occur with repeated detoxifications. Although therefore recognized as an area of medicine that needs new approaches, none have been forthcoming. Indeed, AUD is one of the hardest diagnoses to treat in all of psychiatry.
INCORPORATION BY REFERENCE
[05] Each patent, publication, and non-patent literature cited in the application, or listed in the section entitled "References" below, is hereby incorporated by reference in its entirety as if each was incorporated by reference individually, and as if each is fully set forth herein.
However, where such reference is made, and whether to patents, publications, non-patent literature, or other sources of information, it is for the general purpose of providing context for discussing features of the invention. Accordingly, unless specifically stated otherwise, the reference is not to be construed as an admission that the document or underlying information, in any jurisdiction, is prior art, or forms part of the common general knowledge in the art.
BRIEF SUMMARY OF THE INVENTION
[06] The present invention discloses compositions of MDMA and their use in methods of MDMA-assisted psychotherapy to treat patients with alcohol use disorder. Such compositions and methods have various advantages over current treatments, as further discussed herein.
[07] The following presents a simplified summary of some embodiments of the invention and some of the advantages of the claimed compositions and methods, in order to provide a basic understanding of the invention. This summary is not an extensive overview of the invention, nor intended to identify key or critical elements of the invention or to delineate the scope thereof Its sole purpose is to present some embodiments of the invention in a simplified form as a prelude to the detailed description that follows.
[08] Herein are disclosed methods of treating alcohol use disorder (AUD) in a subject diagnosed therewith, comprising facilitating an MDMA-assisted psychotherapy regimen, wherein the regimen comprises one or more MDMA-assisted psychotherapy sessions and one or more non-drug psychotherapy sessions.
[09] In some embodiments of the invention, the method further comprises an alcohol detoxification course prior to the MDMA-assisted psychotherapy regimen. In some embodiments of the invention, the method further comprises a pre-detoxification motivational group therapy course prior to the alcohol detoxification course. In some embodiments of the invention, the method further comprises a group therapy preparation course prior to the MDMA-assisted psychotherapy regimen.
[10] Herein are disclosed methods of maintaining reduced alcohol intake in a subject diagnosed with AUD, comprising facilitating an MDMA-assisted psychotherapy regimen, wherein the regimen comprises one or more MDMA-assisted psychotherapy sessions and one or more non-drug psychotherapy sessions.
[11] In some embodiments of the invention, the reduced alcohol intake is maintained for at least 1 month after the subject has completed the MDMA-assisted psychotherapy regimen. In some embodiments of the invention, the reduced alcohol intake is maintained for at least 3 months after the subject has completed the MDMA-assisted psychotherapy regimen. In some embodiments of the invention, the reduced alcohol intake is maintained for at least 6 months after the subject has completed the MDMA-assisted psychotherapy regimen. In some embodiments of the invention, the reduced alcohol intake is maintained for at least 9 months after the subject has completed the MDMA-assisted psychotherapy regimen. In some embodiments of the invention, the reduced alcohol intake is maintained for at least 12 months after the subject has completed the MDMA-assisted psychotherapy regimen
[12] In some embodiments of the invention, alcohol intake is reduced by at least 50%. In some embodiments of the invention, alcohol intake is reduced by at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 100%. In some embodiments of the invention, the subject remains completely abstinent from alcohol.
[13] Herein are disclosed methods of preventing relapse into alcohol use in a subject diagnosed with AUD, comprising facilitating an MDMA-assisted psychotherapy regimen, wherein the regimen comprises one or more MDMA-assisted psychotherapy sessions and one or more non-drug psychotherapy sessions.
[14] In some embodiments of the invention, the relapse into alcohol use is prevented for at least 1 month after the subject has completed the 1\4-DMA-assisted psychotherapy regimen. In some embodiments of the invention, the relapse into alcohol use is prevented for at least 3 months after the subject has completed the MDMA-assisted psychotherapy regimen. In some embodiments of the invention, the relapse into alcohol use is prevented for at least 6 months after the subject has completed the MDMA-assisted psychotherapy regimen. In some embodiments of the invention, the relapse into alcohol use is prevented for at least 9 months after the subject has completed the MDMA-assisted psychotherapy regimen. In some embodiments of the invention, the relapse into alcohol use is prevented for at least 12 months after the subject has completed the MDMA-assisted psychotherapy regimen.
[15] Herein are disclosed methods of treating the symptoms of a comorbid psychiatric disorder in a subject diagnosed with AUD, comprising facilitating an MDMA-assisted psychotherapy regimen, wherein the regimen comprises one or more MDMA-assisted psychotherapy sessions and one or more non-drug psychotherapy sessions.
[16] In some embodiments of the invention, the comorbid psychiatric disorder is any of post-traumatic stress disorder (PTSD), adjustment disorder, affective disorder, depression, atypical depression, postpartum depression, catatonic depression, a depressive disorder due to a medical condition, premenstrual dysphoric disorder, seasonal affective disorder, dysthymia, anxiety, phobia disorders, binge disorders, body dysmorphic disorder, a mood disorder related to another health condition, disruptive behavior disorders, eating disorders, impulse control disorders, obsessive compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), personality disorders, attachment disorders, dissociative disorders, and substance-related disorders including substance use disorder (SUD)
[17] In some embodiments of the invention, the comorbid psychiatric disorder is depression or anxiety. In some embodiments of the invention, the comorbid psychiatric disorder is depression, and the symptoms are treated as demonstrated by a reduction in a depression score. In some embodiments of the invention, the comorbid psychiatric disorder is anxiety, and the symptoms are treated as demonstrated by a reduction in a GAD-7 anxiety score.
[18] In some embodiments of the invention, the comorbid psychiatric disorder is a behavioral addiction, including any of pathological gambling, kleptomania, pyromania, compulsive buying, compulsive sexual behaviour, Internet addiction, pornography, and binge eating disorder.
[19] In some embodiments of the invention, the subject has dependent use AUD.
In some embodiments of the invention, the subject has harmful use AUD. In some embodiments of the invention, the subject has alcohol abuse, alcohol dependence, alcohol addiction, or alcoholism.
[20] In some embodiments of the invention, the subject has demonstrated motivation to reduce their alcohol intake. In some embodiments of the invention, the subject has successfully completed the alcohol detoxification course prior to the MDMA-assisted psychotherapy regimen. In some embodiments of the invention, the subject has successfully completed the pre-detoxification motivational group therapy course prior to the alcohol detoxification course.
In some embodiments of the invention, the subject has successfully completed the group therapy preparation course prior to the MDMA-assisted psychotherapy regimen.
[21] In some embodiments of the invention, the subject is diagnosed with AUD
as defined by a version of the Diagnostic and Statistical Manual of Mental Disorders (DSM), comprising scoring above the diagnostic threshold for AUD. In some embodiments of the invention, the severity of AUD is determined using one or more questionnaires. In some embodiments of the invention, the one or more questionnaire(s) comprise the Severity of Alcohol Questionnaire (SADQ) and/or the Short Inventory of Problems for Alcohol (SIP) questionnaire.
In some embodiments of the invention, the one or more questionnaire(s) comprise the Severity of Alcohol Questionnaire (SADQ). In some embodiments of the invention, the severity of AUD
is scored on the SADQ as mild (score of less than 16), moderate (score of 16-30), or severe (score of greater than or equal to 31). In some embodiments of the invention, the severity of AUD is mild (score of less than 16). In some embodiments of the invention, the severity of AUD is moderate (score of 16-30). In some embodiments, the severity of AUD is severe (score of greater than or equal to 31).
[22] In some embodiments of the invention, the method results in one or more of a reduction of alcohol use, a reduction of alcohol cravings, a promotion of alcohol abstinence, a prevention of relapse into alcohol use, or an improvement of at least one symptom of alcohol use disorder.
[23] In some embodiments of the invention, the method results in one or more of an increase in quality of life, an increase in psychosocial functioning, a decrease in use or frequency of a prescription medication, a decrease in use or frequency of a recreational drug, a decrease in obsessive compulsive thoughts, a decrease in suicidality, an increase in feelings of empathy, or an increase in self-compassion.
[24] In some embodiments of the invention, the method results in one or more of an increase in physical functioning, a decrease in role limitations due to physical health, an increase in emotional well-being, a decrease in role limitations due to emotional problems, an increase in energy, a decrease in fatigue, an increase in social functioning, a decrease in pain, or an increase in general health.
[25] In some embodiments of the invention, the increase and/or the decrease is demonstrated by a patient-reported measure on a Short Form (36) Health Survey.
[26] In some embodiments of the invention, the results are measured at least 1 month, at least 3 months, at least 6 months, at least 9 months, or at least 1 year from the end of detoxification, if detoxification is completed, from baseline, from the first psychotherapy session, or from the completion of the MDMA-assisted psychotherapy regimen.
[27] In some embodiments of the invention, the 1\4:DMA-assisted psychotherapy regimen comprises one or more non-drug psychotherapy sessions prior to and after administration of MDMA in conjunction with psychotherapy. In some embodiments of the invention, the MDMA-assisted psychotherapy regimen comprises more than one non-drug psychotherapy session prior to and after the one or more MDMA-assisted psychotherapy sessions. In some embodiments of the invention, the MDMA-assisted psychotherapy regimen comprises administering one or more doses of MDMA during the one or more MDMA-assisted psychotherapy sessions. In some embodiments of the invention, the MDMA-assisted psychotherapy regimen comprises repeated MDMA-assisted psychotherapy sessions.
In some embodiments of the invention, the MDMA-assisted psychotherapy regimen comprises an initial MDMA-assisted psychotherapy session and a subsequent MDMA-assisted psychotherapy session. In some embodiments of the invention, the MDMA-assisted psychotherapy regimen comprises one, two, three, or four non-drug psychotherapy sessions prior to a first dose of MDMA in conjunction with psychotherapy. In some embodiments of the invention, the MDMA-assisted psychotherapy regimen comprises two non-drug psychotherapy sessions prior to a first MDMA-assisted psychotherapy session.
In some embodiments of the invention, the MDMA-assisted psychotherapy regimen comprises one or more non-drug psychotherapy session(s) after the first MDMA-assisted psychotherapy session.
In some embodiments of the invention, the MDMA-assisted psychotherapy regimen comprises one, two, three, four, five, or six non-drug psychotherapy sessions after the first MDMA-assisted psychotherapy session and prior to a subsequent MDMA-assisted psychotherapy session. In some embodiments of the invention, the MDMA-assisted psychotherapy regimen comprises three non-drug psychotherapy sessions after the first MDMA-assisted psychotherapy session and prior to the subsequent MDMA-assisted psychotherapy session. In some embodiments of the invention, the subsequent MDMA-assisted psychotherapy session is a second, a third, a fourth, or a fifth MDMA-assisted psychotherapy session.
In some embodiments of the invention, the subsequent MDMA-assisted psychotherapy session is a second MDMA-assisted psychotherapy session. In some embodiments of the invention, the MDMA-assisted psychotherapy regimen comprises two MDMA-assisted psychotherapy sessions and eight non-drug psychotherapy sessions. In some embodiments of the invention, the MDMA-assisted psychotherapy regimens comprises three MDMA-assisted psychotherapy sessions and 11 psychotherapy non-drug sessions.
[28] In some embodiments of the invention, the one or more MDMA-assisted psychotherapy sessions comprise administering one single dose of MDMA. In some embodiments of the invention, the first and the subsequent MDMA-assisted psychotherapy sessions each comprise administering one single dose of MDMA. In some embodiments of the invention, the one or more MDMA-assisted psychotherapy sessions comprise administering two doses of MDMA, an initial dose and a booster dose. In some embodiments of the invention, the first and the subsequent MDMA-assisted psychotherapy sessions each comprise two doses of MDMA, an initial dose and a booster dose. In some embodiments of the invention, the booster dose is less than the initial dose. In some embodiments of the invention, the booster dose is about 10% to about 90% of the amount of the initial dose. In some embodiments of the invention, the booster dose is about 25%, about 50%, or about 75% of the amount of the initial dose.
In some embodiments of the invention, the booster dose is about 10%, 15%, 20%, 25%, 30%, 35%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, or 90% of the amount of the initial dose. In some embodiments of the invention, the booster dose is administered about 30 minutes, one hour, two hours, three hours, or four hours after the initial dose. In some embodiments of the invention, the booster dose is administered two hours following the initial dose, or at such time as to extend peak subjective effects or to optimize the plasma concentration-time profile.
[29] In some embodiments of the invention, the MDMA-assisted psychotherapy regimen comprises between 5 and 20 psychotherapy sessions, inclusive, during which at least two of said psychotherapy sessions the subject is administered MDMA. In some embodiments of the invention, a total dose of about 40 mg to 200 mg of MDMA is administered, wherein the range is inclusive. In some embodiments of the invention, a total dose of about 40 mg to 100 mg, about 100 mg to 150 mg, about 150 to 200 mg, about 175 mg to 200 mg, about 180 mg to 190 mg, about 180 to 200 mg, or about 185 to 200 mg of MDMA is administered, wherein each range is inclusive. In some embodiments of the invention, a total dose of about 180 mg, 181 mg, 182 mg, 183 mg, 184 mg, 185 mg, 186 mg, 187 mg, 188 mg, 189 mg, or 190 mg of MDMA
is administered. In some embodiments of the invention, a total dose of about 187.5 mg of MDMA is administered.
[30] In some embodiments of the invention, MDMA is administered as a pharmaceutically acceptable salt, as a free base, or as a polymorph of MDMA. In some embodiments of the invention, MDMA is administered as a racemate. In some embodiments of the invention, MDMA is administered as an enantiomerically enriched mixture of R-MDMA and S-MDMA
having an enantiomeric excess in any amount greater than 0%. In some embodiments of the invention, MDMA is administered as a pure or substantially pure individual enantiomer of R-MDMA or S-MDMA having an enantiomeric excess greater than 90%.
[31] In some embodiments of the invention, the psychotherapy in MDMA-assisted and non-drug sessions comprises motivational interviewing. In some embodiments of the invention, the psychotherapy in MDMA-assisted and non-drug sessions comprises cognitive behavioral therapy. In some embodiments of the invention, the psychotherapy in MDMA-assisted and non-drug sessions comprises motivational interviewing and cognitive behavioral therapy. In some embodiments of the invention, the cognitive behavioral therapy is a modified form of cognitive behavioral therapy comprising elements of Acceptance and Commitment Therapy (ACT).
[32] In some embodiments of the invention, the MDMA-assisted and drug-free psychotherapy sessions comprise one or more of supporting subjects to maintain and/or improve a reduction in alcohol consumption, preventing relapse into heavy drinking, and developing skills to lead a fulfilling life without problematic drinking.
[33] Herein are disclosed methods of selecting a subject for MDMA-assisted psychotherapy, comprising identifying a subject who meets one or more of the following criteria: has an AUD
diagnosis, wherein the severity of AUD is mild, moderate, or severe; has completed detoxification of alcohol prior to administration of MDMA-assisted psychotherapy; and has tapered off of medications known to interact with or attenuate the effects of MDMA prior to administration of MDMA-assisted psychotherapy; wherein the subject does not experience adverse events associated with administration of MDMA.
[34] Herein are disclosed methods of minimizing or eliminating the risk of adverse events related to MDMA-assisted psychotherapy in a subject having AUD comprising:
selecting a subject who has been diagnosed with AUD, wherein the severity of AUD is mild, moderate, or severe; facilitating detoxification of alcohol prior to administration of MDMA-assisted psychotherapy; and tapering off medications known to interact with or attenuate the effects of MDMA prior to administration of MDMA-assisted psychotherapy; wherein the subject does not experience adverse events associated with administration of MDMA.
[35] In some embodiments of the invention, detoxification comprises gradually cutting down alcohol consumption over many weeks and/or is a medically assisted detoxification regime. In some embodiments of the invention, the method further comprises facilitating a pre-detoxification motivational group therapy course prior to facilitating the detoxification of alcohol.
[36] Herein are disclosed methods of selecting a subject for MDMA-assisted psychotherapy, comprising identifying a subject who meets one or more of the following criteria: has an AUD
diagnosis, wherein the severity of AUD is mild, moderate, or severe; has completed a group therapy preparation course prior to administration of MDMA-assisted psychotherapy; and has tapered off of medications known to interact with or attenuate the effects of MDMA prior to administration of MDMA-assisted psychotherapy; wherein the subject does not experience adverse events associated with administration of MDMA.
[37] In some embodiments of the invention, the methods comprise monitoring the subject having AUD for adverse events during administration of MDMA. In some embodiments of the invention, the adverse events during administration of MDMA comprise one or more of hyperthermia, hypertension, and elevated heart rate. In some embodiments of the invention, the subject diagnosed with AUD does not display adverse events during administration of MDMA.
[38] In some embodiments of the invention, the methods comprise monitoring the subject having AUD for adverse events after administration of MDMA. In some embodiments of the invention, the adverse events after administration of MDMA comprise mood disturbances and/or depressed affect. In some embodiments of the invention, the subject does not report a disturbance in mood or a depressed affect after administration of MDMA-assisted psychotherapy. In some embodiments of the invention, the subject does not report a disturbance in mood or a depressed affect within about one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, or ten days after administration.
In some embodiments of the invention, the subject diagnosed with AUD does not display adverse events after administration of MDMA-assisted psychotherapy.
[39] In some embodiments of the invention, the methods comprise monitoring the subject having AUD for adverse events both during and after administration of MDMA. In some embodiments of the invention, the adverse events during administration of MDMA
comprise one or more of hyperthermia, hypertension, and elevated heart rate, and the adverse events after administration of MDMA comprise mood disturbances and/or depressed affect. In some embodiments of the invention, the subject diagnosed with AUD does not display or report adverse events during or after administration of MDMA-assisted psychotherapy.
In some embodiments of the invention, the subject does not display adverse events during administration of MDMA, or report adverse events after administration of MDMA, including within about one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, or ten days after administration.
[40] In some embodiments of the invention, the methods comprise evaluating the severity of anxiety and/or depression in a subject diagnosed with AUD prior to MDMA-assisted psychotherapy. In some embodiments of the invention, the severity of anxiety and/or depression are assessed using questionnaires. In some embodiments of the invention, the severity of anxiety and/or depression are assessed using the Generalised Anxiety Disorder Assessment (GAD-7) and/or the Patient Health Questionnaire-9 (PHQ-9) questionnaire(s). In some embodiments of the invention, the severity of anxiety and/or depression are also assessed after MDMA-assisted psychotherapy, and are reduced after the MDMA-assisted psychotherapy in comparison to levels preceding said MDMA-assisted psychotherapy.
[41] In some embodiments of the invention, AUD is diagnosed as defined by a version of the Diagnostic and Statistical Manual of Mental Disorders (DSM), wherein diagnosis comprises scoring above the diagnostic threshold for AUD. In some embodiments of the invention, the subject has dependent use AUD. In some embodiments of the invention, the subject has harmful use AUD. In some embodiments of the invention, the subject has alcohol abuse, alcohol dependence, alcohol addiction, or alcoholism.
[42] In some embodiments are disclosed a method of treating an alcohol use disorder (AUD) subject, the method comprising: confirming the subject has alcohol use disorder; conducting a screening visit; confirming the subject meets all inclusion criteria;
confirming the subject does not meet any exclusion criteria; conducting a plurality of psychotherapy sessions as part of a treatment protocol, wherein at least three of the plurality of psychotherapy sessions include at least one dose of MDMA (MDMA-assisted psychotherapy sessions); and evaluation of results at predetermined intervals after the subject completes the treatment protocol.
[43] In some embodiments of the invention, the treatment protocol includes between 5 and 20 sessions or between 10 and 15 sessions. In some embodiments of the invention, the treatment protocol includes 14 sessions. In some embodiments of the invention, the treatment protocol includes 3 MDMA-assisted psychotherapy sessions, and 11 non-drug psychotherapy sessions. In some embodiments of the invention, the MDMA-assisted psychotherapy sessions comprise at least one MDMA administration.In some embodiments of the invention, the at least one MDMA administration comprises an initial dose and optionally a booster dose. In some embodiments of the invention, the initial dose is between about 100 mg to about 150 mg, and wherein the optional booster dose is half of the initial dose. In some embodiments of the invention, the optional booster dose is administered between about 90 minutes to about 120 minutes after the initial dose. In some embodiments of the invention, the third, seventh, and eleventh sessions are MDMA-assisted psychotherapy sessions. In some embodiments of the invention, the MDMA-assisted psychotherapy sessions last from between about 360 minutes to about 480 minutes. In some embodiments of the invention, the evaluation of results occurs at 3 months, 6 months, and 9 months after a subject successfully completes the treatment protocol.
[44] In some embodiments of the invention, prior to conducting a plurality of psychotherapy sessions, the subject completes a group therapy preparation course. In some embodiments of the invention, the group therapy preparation course includes a plurality of sessions. In some embodiments of the invention, each of the plurality of sessions last for about 60 minutes. In some embodiments of the invention, each of the plurality of sessions includes a discussion surrounding a specific topic. In some embodiments of the invention, the plurality of sessions includes a first session, a second session, a third session, and a fourth session. In some embodiments of the invention, the specific topic of the first session is building confidence; the specific topic of the second session is resources, barriers, and goal setting;
the specific topic of the third session is increasing rewarding activities and building support; the specific topic of the fourth session is building the foundations for recovery. In some embodiments of the invention, prior to conducting a plurality of psychotherapy sessions, the subject completes a detoxification course.
[45] Herein are disclosed methods of verifiably reducing alcohol consumption by a subject diagnosed with alcohol use disorder (AUD), comprising the steps of:
determining the amount of alcohol consumed by the subject prior to completing the method; conducting a treatment protocol comprising 14 psychotherapy sessions, wherein 3 of the 14 psychotherapy sessions are MDM A -as si sted psychotherapy sessions, and each of the MDMA -as si sted psychotherapy sessions includes an initial dose, and optionally a booster dose, wherein the initial dose is between about 100 mg to about 150 mg, and the optional booster dose is about half of the initial dose, measuring the amount of alcohol consumed of the subject three months, six months, and nine months after the final psychotherapy session; verifying that the amount of alcohol consumed by the subject nine months after the final psychotherapy session is less than the amount consumed by the subject as determined prior to completing the method.
[46] In some embodiments of the invention, the amount of alcohol consumed by the subject nine months after the final psychotherapy session is at least 50% less than the amount consumed by the subject as determined prior to completing the method. In some embodiments of the invention, the amount of alcohol consumed by the subject nine months after the final psychotherapy session is at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% less than the amount consumed by the subject as determined prior to completing the method
[47] Herein are disclosed methods of decreasing the daily alcohol consumption in a subject diagnosed with alcohol use disorder (AUD), the method comprising: completing an 11 week psychotherapy regimen comprising 14 psychotherapy sessions, wherein 3 of the psychotherapy sessions are about 360 minute to about 480 minute long MDMA-assisted psychotherapy sessions comprising the administration of two doses of MDMA, the two doses including an initial dose and a booster dose, the initial dose comprising between about 100 mg to about 150 mg of MDMA, and the booster dose, administered between about 90 minutes to about 120 minutes after the initial dose, comprising about half of the initial dose, and wherein 11 of the 14 psychotherapy sessions are non-drug psychotherapy sessions lasting about 60 minutes. In some embodiments of the invention, the MDMA-assisted psychotherapy sessions occur once every 3 weeks for 9 weeks of the psychotherapy regimen, and the non-drug psychotherapy sessions occur once every week for all 11 weeks of the psychotherapy regimen, so that a non-dosing psychotherapy session follows an MDMA dosing session on weeks where the MDMA dosing session occurs.
[48] Herein are disclosed methods of treating alcohol use disorder (AUD) in a subject diagnosed therewith comprising a psychotherapy treatment protocol comprising the steps of:
conducting a screening visit, wherein the screening visit confirms the subject meets all inclusion criteria and does not meet any exclusion criteria; completion of a plurality of psychotherapy sessions as part of a treatment protocol conducted under the supervision of a dyadic therapy team, the plurality of psychotherapy sessions including: a first session, wherein the dyadic therapy pair introduces the therapy program to the subject, prepares the subject for the treatment protocol, and discusses the subject's personal story, a second session, wherein the dyadic therapy team teaches and practices a relaxation technique with the subject and reviews the subject's current recovery plan; a third session, wherein the subject is administered at least one dose of MDMA, wherein the at least one dose of MDMA is between about 100 mg to about 150 mg; a fourth session, wherein the dyadic therapist team guides the subject through a mindfulness routine, and the subject optionally provides a narrative of their experience after being administered at least one dose of MDMA; a fifth session, wherein the dyadic therapy team discusses high risk situations that may lead to a relapse, and the workability of alcohol as a control strategy, with the subject; a sixth session, wherein the dyadic therapist team practices at least one defusion technique with the subject; a seventh session, wherein the subject is administered at least one dose of MDMA, wherein the at least one dose of MDMA
is between about 100 mg to about 150 mg; an eighth session, wherein the dyadic therapist team guides the subject through a mindfulness routine, and the subject optionally provides a narrative of their experience after being administered at least one dose of MDMA; a ninth session, wherein the dyadic therapist team aids the subject in identifying problematic avoidance of internal experiences and practices at least one acceptance technique with the subject;
a tenth session, wherein the dyadic therapist team aids the subject in identifying at least one thing important in their life, at least one skill they possess, and at least one barrier that may impact their success;
an eleventh session, wherein the subject is administered at least one dose of MDMA, wherein the at least one dose of MDMA is between about 100 mg to about 150 mg; a twelfth session, wherein the dyadic therapist team guides the subject through a mindfulness routine, and the subject optionally provides a narrative of the MDMA administration; a thirteenth session, wherein the dyadic therapist team aids the subject in developing self-compassion, recognizing urges to consume alcohol, and practicing methods of establishing everyday mindfulness; and a fourteenth session, wherein the dyadic therapist team reviews the treatment protocol with the subject, and establishes an ongoing recovery plan with the subject.
[49] Herein is disclosed 1VIDMA for use in a method of treating alcohol use disorder (AUD) in a subject diagnosed therewith, comprising a psychotherapy treatment protocol comprising the steps of: conducting a screening visit, wherein the screening visit confirms the subject meets all inclusion criteria and does not meet at least one exclusion criteria;
completion of a plurality of psychotherapy sessions as part of a treatment protocol conducted under the supervision of a dyadic therapy team, the plurality of psychotherapy sessions including: a first session, wherein the dyadic therapy pair introduces the therapy program to the subject, prepares the subject for the treatment protocol, and discusses the subject's personal story, a second session, wherein the dyadic therapy team teaches and practices a relaxation technique with the subject and reviews the subject's current recovery plan; a third session, wherein the subject is administered at least one dose of MDMA, wherein the at least one dose of MDMA is between about 100 mg to about 150 mg; a fourth session, wherein the dyadic therapist team guides the subject through a mindfulness routine, and the subject optionally provides a narrative of their experience after being administered at least one dose of MDMA; a fifth session, wherein the dyadic therapy team discusses high risk situations that may lead to a relapse, and the workability of alcohol as a control strategy, with the subject; a sixth session, wherein the dyadic therapist team practices at least one defusion technique with the subject; a seventh session, wherein the subject is administered at least one dose of MDMA, wherein the at least one dose of MDMA
is between about 100 mg to about 150 mg; an eighth session, wherein the dyadic therapist team guides the subject through a mindfulness routine, and the subject optionally provides a narrative of their experience after being administered at least one dose of MDMA; a ninth session, wherein the dyadic therapist team aids the subject in identifying problematic avoidance of internal experiences and practices at least one acceptance technique with the subject;
a tenth session, wherein the dyadic therapist team aids the subject in identifying at least one thing important in their life, at least one skill they possess, and at least one barrier that may impact their success;
an eleventh session, wherein the subject is administered at least one dose of MDMA, wherein the at least one dose of MDMA is between about 100 mg to about 150 mg; a twelfth session, wherein the dyadic therapist team guides the subject through a mindfulness routine, and the subject optionally provides a narrative of the MDMA administration; a thirteenth session, wherein the dyadic therapist team aids the subject in developing self-compassion, recognizing urges to consume alcohol, and practicing methods of establishing everyday mindfulness; and a fourteenth session, wherein the dyadic therapist team reviews the treatment protocol with the subject, and establishes an ongoing recovery plan with the subject.
[50] Herein are disclosed methods of reducing the units of alcohol consumed per week in a subject diagnosed with alcohol use disorder (AUD) by between about 70% to about 100% after completing the method, the method comprising. determining the amount of alcohol consumed by the subject in units per week prior to completing the method; conducting a treatment protocol comprising 14 psychotherapy sessions, wherein 3 of the 14 psychotherapy sessions are MDMA-assisted psychotherapy sessions, and each of the MDMA-assisted psychotherapy sessions includes an initial dose, and optionally a booster dose, wherein the initial dose is between about 100 mg to about 150 mg, and the optional booster dose is about half of the initial dose; measuring the amount of alcohol consumed of the subject in units per week three months, six months, and nine months after completing the 14 psychotherapy sessions;
verifying that the amount of alcohol consumed by the subject nine months after completing the 14 psychotherapy sessions is between about 70% to about 100% less than the amount consumed by the subject as determined prior to completing the method.
[51] In some embodiments of the invention, the subject has a genetic variation associated with a substance use disorder, such as alcohol use disorder, and including a genetic variation in mGluR5 or or FKBP5.
[52] In some embodiments of the invention, the subject is administered one or more therapeutic agents in addition to MDMA. In some embodiments of the invention, the subject is also administered a therapeutically effective amount of an additional active agent selected from the group consisting of: an opioid antagonist (e.g., nalmefene, naltrexone), a CB1 antagonist (e.g., rimonabant), a CRH1 receptor antagonist (e.g., verucerfont, pexacerfont), a NK1R antagonist (e.g., tradipitant), an OTR agonist (e.g., oxytocin), a GABA
agent (e.g., topiramate, baclofen, a benzodiazepine, such as alprazolam, diazepam or lorazepam), a voltage-gated sodium channel inhibitor (e.g., oxacarbazepine, valproic acid, zonisamide), a voltage-dependent calcium channel agonist (e.g., gabapentin, pregabalin), an a7 nicotinic acetylcholine receptor agonist (e.g., varenicline), a 5-HT3 antagonist (e.g., ondansetron), a 5-HT1A, receptor partial agonist (e.g., aripiprazole), a 5-HT2A receptor antagonist (e.g., quetiapine, olanzapine, mirtazapine), a 5-HT reuptake inhibitor (e.g., trazodone), a SERT
inhibitor (e.g., duloxetine), a al adrenoreceptor antagonist (e.g., doxazosin, prazosin), a glucocorticoid receptor antagonist (e.g., mifepristone), an al adrenoreceptor agonist (e.g., guanfacine), an AChE inhibitor (e.g., citicoline), a dopamine D2 receptor antagonist (e.g., tiapride), an a2 adrenoreceptor agonist (e.g., clonidine), an NMDA receptor antagonist (e.g., acamprosate) and an aldehyde dehydrogenase inhibitor (e.g., disulfiram), and pharmaceutically acceptable salts thereof
[53] Disclosed herein are pharmaceutical compositions of MDMA for use in the method of the invention, wherein said composition comprises a substantially pure MDMA
compound having a chromatographic purity of greater than 95% as measured by HPLC. In some embodiments of the invention, the substantially pure MDMA is substantially free of impurities or adulterants.
[54] Disclosed herein are pharmaceutical compositions of MDMA for use in the method of the invention which is a pure or substantially pure individual enantiomer of R-MDMA or S-MDMA having an enantiomeric excess greater than 90%, or an enantiomerically enriched mixture having an enantiomeric excess in any amount greater than 0%.
[55] Disclosed herein are pharmaceutical compositions of MDMA for use in the method of the invention, wherein said MDMA is in unit dosage form. In some embodiments of the invention, the unit dosage form is an immediate release, controlled release, sustained release, extended release, or modified release formulation.
[56] Disclosed herein is MDMA for use in a method of reducing the units of alcohol consumed per week in a subject diagnosed with alcohol use disorder (AUD) by between about 70% to about 100% after completing the method, the method comprising.
determining the amount of alcohol consumed by the subject in units per week prior to completing the method;
completion by the subject of a preparation group course; conducting a treatment protocol comprising 14 psychotherapy sessions, wherein 3 of the 14 psychotherapy sessions are MDMA-assisted psychotherapy sessions, and each of the MDMA-assisted psychotherapy sessions includes an initial dose, and optionally a booster dose, wherein the initial dose is between about 100 mg to about 150 mg, and the optional booster dose is about half of the initial dose; measuring the amount of alcohol consumed of the subject in units per week three months, six months, and nine months after completing the 14 psychotherapy sessions;
verifying that the amount of alcohol consumed by the subject nine months after completing the 14 psychotherapy sessions is between about 70% to about 100% less than the amount consumed by the subject as determined prior to completing the method.
[57] Disclosed herein is MDMA for use in a method of decreasing the daily alcohol consumption in a subject diagnosed with alcohol use disorder, the method comprising:
completing an 11 week psychotherapy regimen comprising 14 psychotherapy sessions, wherein 3 of the 14 psychotherapy sessions are about 360 minute to about 480 minute long MDMA-assisted psychotherapy sessions comprising the administration of two doses of MDMA, the two doses including an initial dose and a booster dose, the initial dose comprising between about 100 mg to about 150 mg of MDMA, and the booster dose, administered between about 90 minutes to about 120 minutes after the initial dose, comprising about half of the initial dose, and wherein 11 of the 14 psychotherapy sessions are non-drug psychotherapy sessions lasting about 60 minutes.
[58] Disclosed herein are uses of MDMA for the manufacture of a medicament for a treatment for an alcohol use disorder (AUD) subject, according to the methods of the invention.
In some embodiments of the invention, the AUD subject has dependent use AUD, harmful use AUD, alcohol abuse, alcohol dependence, alcohol addiction, or alcoholism.
[59] Disclosed herein are uses of MDMA in conjunction with psychotherapy to treat an alcohol use disorder (AUD) subject, according to the methods of the invention.
Disclosed herein are uses of MDMA for the manufacture of a medicament for a treatment to reduce alcohol use by an AUD subject, according to the methods of the invention.
Disclosed herein are uses of MDMA for the manufacture of a medicament for a treatment to prevent relapse into alcohol use by an AUD subject, according to the methods of the invention.
Disclosed herein are uses of MDMA for the manufacture of a medicament for a treatment to promote alcohol abstinence by an AUD subject, according to the methods of the invention.
[60] Disclosed herein are uses of MDMA for the manufacture of a medicament for the treatment of the symptoms of depression or anxiety associated with AUD, according to the methods of the invention. Disclosed herein are uses of MDMA for the manufacture of a medicament for the treatment of the symptoms of a psychiatric disorder comorbid with AUD, according to the methods of the invention. In some embodiments of the invention, the psychiatric disorder comorbid with AUD is any of post-traumatic stress disorder (PTSD), adjustment disorder, affective disorder, depression, atypical depression, postpartum depression, catatonic depression, a depressive disorder due to a medical condition, premenstrual dysphoric disorder, seasonal affective disorder, dysthymia, anxiety, phobia disorders, binge disorders, body dysmorphic disorder, a mood disorder related to another health condition, disruptive behavior disorders, eating disorders, impulse control disorders, obsessive compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), personality disorders, attachment disorders, dissociative disorders, or substance-related disorders including substance use disorder (SUD). In some embodiments of the invention, the psychiatric disorder comorbid with AUD is a behavioral addiction, including any of pathological gambling, kleptomania, pyromania, compulsive buying, compulsive sexual behaviour, Internet addiction, pornography, and binge eating disorder.
[61] In some embodiments of the invention, the use of MDMA comprises monitoring adverse events during MDMA-assisted psychotherapy. In some embodiments of the invention, the adverse events comprise one or more of hyperthermia, hypertension, and elevated heart rate. In some embodiments of the invention, adverse events are not observed during MDMA-assisted psychotherapy.
[62] In some embodiments of the invention, the use of MDMA comprises monitoring for adverse events after administration of MDMA-assisted psychotherapy. In some embodiments of the invention, the adverse events after MDMA-assisted psychotherapy comprise mood disturbances and/or depressed affect. In some embodiments of the invention, a mood disturbance and/or depressed affect after administration of MDMA-assisted psychotherapy are not observed and/or reported. In some embodiments of the invention, mood disturbances and/or depressed affect are not observed and/or reported within about one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, or ten days after MDMA-assisted psychotherapy. In some embodiments of the invention, adverse events after administration of MDMA-assisted psychotherapy are not observed or reported.
[63] In some embodiments of the invention, the use of MDMA comprises monitoring for adverse events both during and after administration of MDMA.In some embodiments of the invention, the adverse events during administration of MDMA comprise one or more of hyperthermia, hypertension, and elevated heart rate, and the adverse events after administration of MDMA comprise mood disturbances and/or depressed affect. In some embodiments of the invention, no adverse events are observed or reported during or after administration of MDMA-assisted psychotherapy. In some embodiments of the invention, no adverse events are observed or reported during administration of MDMA, or reported after administration of MDMA, including within about one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, or ten days after administration.
[64] In some embodiments of the invention, the subject has a genetic variation associated with a substance use disorder, such as alcohol use disorder, and including a genetic variation in mGluR5 or or FKBP5.
[65] In some embodiments of the invention, the subject is administered one or more therapeutic agents in addition to MDMA. In some embodiments of the invention, the subject is al so administered a therapeutically effective amount of an additional active agent selected from the group consisting of: an opioid antagonist (e.g., nalmefene, naltrexone), a CB1 antagonist (e.g., rimonabant), a CRH1 receptor antagonist (e.g., verucerfont, pexacerfont), a NK1R antagonist (e.g., tradipitant), an OTR agonist (e.g., oxytocin), a GABA
agent (e.g., topiramate, baclofen, a benzodiazepine, such as alprazolam, diazepam or lorazepam), a voltage-gated sodium channel inhibitor (e.g., oxacarbazepine, valproic acid, zonisamide), a voltage-dependent calcium channel agonist (e.g., gabapentin, pregabalin), an a7 nicotinic acetylcholine receptor agonist (e.g., varenicline), a 5-HT3 antagonist (e.g., ondansetron), a 5-HT1A, receptor partial agonist (e.g., aripiprazole), a 5-HT2A receptor antagonist (e.g., quetiapine, olanzapine, mirtazapine), a 5-HT reuptake inhibitor (e.g., trazodone), a SERT
inhibitor (e.g., duloxetine), a al adrenoreceptor antagonist (e.g., doxazosin, prazosin), a glucocorticoid receptor antagonist (e.g., mifepristone), an al adrenoreceptor agonist (e.g., guanfacine), an AChE inhibitor (e.g., citicoline), a dopamine D2 receptor antagonist (e.g., tiapride), an a2 adrenoreceptor agonist (e.g., clonidine), an NMDA receptor antagonist (e.g., acamprosate) and an aldehyde dehydrogenase inhibitor (e.g., disulfiram), and pharmaceutically acceptable salts thereof.
[66] The foregoing has outlined broadly and in summary certain pertinent features of the disclosure so that the detailed description of the invention that follows may be better understood, and so that the present contribution to the art can be more fully appreciated. Hence, this summary is to be considered as a brief and general synopsis of only some of the objects and embodiments disclosed herein, is provided solely for the benefit and convenience of the reader, and is not intended to limit in any manner the scope, or range of equivalents, to which the claims are lawfully entitled. Additional features of the invention will be described hereinafter. It should be appreciated by those in the art that all such disclosed specific compositions and methods are only exemplary, and may be readily utilized as a basis for modifying or designing other compositions and methods for carrying out the same purposes.
Such equivalent compositions and methods will be appreciated to be also within the scope and spirit of the invention as set forth in the claims. It also will be appreciated that the headings within this document are being utilized only to expedite its review by a reader. They should not be construed as limiting the invention in any manner.
BRIEF SUMMARY OF THE DRAWINGS
[67] To further clarify various aspects of the present invention, a more particular description of the invention will be rendered by reference to certain exemplary aspects thereof which are illustrated in the included figures. It should be understood and appreciated that the figures depict only illustrated aspects of certain exemplary embodiments of the invention and therefore are not to be considered limiting of its scope. They are simply provided as qualitative illustrations of the concepts of the present invention
[68] Certain aspects of the present invention are therefore further described and explained with additional specificity and detail, but still by way of example only, with reference to the accompanying figures in which:
[69] FIG. 1 illustrates drinking behavior outcomes for patients undergoing a community alcohol detoxification (as measured by TLFB) up to nine months (reproduced from Sessa et al.
2020). Specifically, individual drinking behavior is displayed as units of alcohol consumed per week (n=12). In the case of additional detoxification TLFB results are expressed as an extrapolation of drinking behavior from the point of their initial relapse (dotted lines indicate the point). Incomplete lines indicate drop-out. This population of patients ("Outcomes patients") did not receive MDMA-assisted therapy.
[70] FIG. 2 illustrates drinking behavior outcomes for the Outcomes patients undergoing a community alcohol detoxification (as measured by TLFB) up to nine months (reproduced from Sessa et al. 2020). Specifically, the average weekly alcohol consumption for all 12 patients up to 9 months post-detox as shown in FIG. 1 is illustrated.
[71] FIG. 3 illustrates the results of the PHQ-9 assessment of outcome measures (see TABLE 2 and discussion below), in table and graph form, demonstrating high levels of depression among the population of Outcomes patients, levels of depression that remained significantly high throughout the 9 months of the study (reproduced from Sessa et al. 2020)
[72] FIG. 4 illustrates the results of the PHQ-9 assessment of outcome measures (see TABLE 2 and discussion below), in table and graph form, demonstrating high levels of anxiety among the population of Outcomes patients, levels of anxiety that remained significantly high throughout the 9 months of the study (reproduced from Sessa et al. 2020).
[73] FIG. 5 illustrates the results of the SF-36 outcome measure (see TABLE 2 and discussion below), in table and graph form, suggesting that for those who provided results to 9 months, despite the majority of patients relapsing to high levels of drinking by 3 months (and continuing high rates of drinking, even if they had second or third detoxes) their overall level of psychosocial functioning appears to have improved over the course of 9 months (although an incomplete dataset at 9 months is noted) (reproduced from Sessa et al.
2020).
[74] FIG. 6 illustrates the results of the Severity of Alcohol Dependence Questionnaire (SAD-Q) measure of alcohol dependency, from the patients who received MDMA-assisted psychotherapy as in Example 8. A score of 31 or higher indicates severe alcohol dependence, a score of 16-30 indicates moderate dependence (light grey area), anything lower than 16 indicates mild dependence (dark grey area). Also illustrated are the results of the Short Inventory of Problems for Alcohol (SIP), a 15 question measure of self-noted consequences of drinking. Both observed at screening. Four SAD-Q questions were unanswered, in which case, mean substitution was applied using the average row value for the relevant time period and participant. B05, B16, B20 and B21 had one question missing each.
[75] FIG. 7 illustrates pooled data of Blood Pressure, Temperature, Heart Rate, Observed Drug Effects, and Subjective Units of Distress (SUDS) observed over the duration of the MDMA sessions in patients who underwent MDMA-assisted psychotherapy in the study of Example 8. SUDS and Drug Effects observed over 8 hours, physiological data observed over 6 hours following dosing. Mean data for each session is used, except in the case of missing data, where available session data is applied. Error bars (where applied) indicate +/- SEM.
[76] FIG. 8 illustrates the results of the Timeline Follow Back (TLFB), which assesses drinking behavior prior to, and following the study of Example 8. Data is collected daily by self-reporting and reviewed at one month prior to detox, immediately following detox and at 1, 3, 6 and 9 months follow-up. A full dataset was not available for three of the participants.
One participant dropped out of the study at three months and two patients failed to provide data at the nine month follow-up. Two participants had a second detox since starting the study. For these participants, TLFB drinking behavior data was carried forward from the point of drinking levels pre-second detox.
[77] FIG. 9 illustrates the results of the Profile of Mood States (POMS), individual composite scores of mood disturbance observed daily over a week following dosing, obtained from the patients in the study of Example 8. Mean data shown for both MDMA
sessions. In the case of absent data for either session the available data for the remaining session is used.
[78] FIG. 10 illustrates the results of General Anxiety Disorder 7 (GAD-7) and Patient Health Questionnaire 9 (PHQ-9) obtained from the patients in the study of Example 8 These are self-report scales for anxiety and depression, respectively (as shown for Outcomes patients in FIGS. 3 and 4). Recording was at screening, baseline, week 10, then 3, 6 and 9 months follow-up. Greater scores report indication of heightened anxiety/depression.
Error bars indicate +/- SEM.
[79] FIG. 11 illustrates TLFB showing the percentage of patients consuming more than the 14 recommended daily units of alcohol, and comparing the results of the patients in the study of Example 8 with the results of the Outcomes patients reported in Sessa et al. 2020.
[80] FIG. 12 illustrates an embodiment of the invention comprising a therapy protocol useful in the treatment of non-physically dependent (harmful use) AUD.
[81] FIG. 13 illustrates an embodiment of the invention comprising a therapy protocol useful in the treatment of physically dependent (dependent use) AUD.
[82] FIG. 14 illustrates an exemplary detoxification course, according to some embodiments of the invention.
[83] FIG. 15 illustrates an overview of an exemplary treatment protocol useful in treating a patient diagnosed with AUD, according to some embodiments of the invention.
[84] FIG. 16 illustrates an exemplary treatment protocol useful in treating a patient diagnosed with AUD, according to some embodiments of the invention.
[85] FIG. 17: illustrates the study procedure for the 13-week study course, including assessments completed, according to some embodiments of the invention.
DETAILED DESCRIPTION OF THE INVENTION
[86] Alcohol use disorder (AUD) (DSM-5, APA 2013), which includes what are sometimes referred to as alcohol abuse, alcohol dependence, alcohol addiction, and colloquially "alcoholism," is a psychiatric disorder in which individuals present with a cluster of symptoms of varying severity, that cause psychological and physical harms and social dysfunction due to the use of alcohol. Although drinking alcohol is a widely socially acceptable behavior and many people consume alcohol without experiencing any problems, a growing number of people drink in a harmful manner. Approximately 24% of the adult population of England consume alcohol in a way that is potentially or actually harmful, i.e., "harmful use"
(McManus et at 2009).
[87] Physical dependence to alcohol ("alcohol dependence" or "dependent use") is a less common, more severe form of AUD. In the UK, the prevalence of physical alcohol dependence is about 4%, with 6% of men and 2% of women meeting criteria for this psychiatric disorder.
Typically, alcohol dependence is characterized by: often serious withdrawal symptoms on cessation of alcohol, or drinking to avoid withdrawal symptoms, tolerance and the persistent desire to drink, and continuing drinking despite negative consequences (NICE
Guidelines on Alcohol Use Disorders, 2011).
[88] "Harmful use" of alcohol, by contrast, is defined in the World Health Organization (WHO)'s International Classification of Diseases, 10th Revision (The ICD-10 Classification of Mental and Behavioural Disorders) as: "[A] pattern of psychoactive substance use that is causing damage to health. The damage may be physical (e.g., hepatitis) or mental (e.g., depressive episodes secondary to heavy alcohol intake). Harmful use commonly, but not invariably, has adverse social consequences; social consequences in themselves, however, are not sufficient to justify a diagnosis of harmful use" (ICD-10; WHO, 1992).
[89] From a clinical perspective, it can be helpful to separate this group of "harmful use"
drinkers from "dependent use" drinkers with more severe and enduring physical dependence upon alcohol that may require medical support (e.g., detoxification with benzodiazepine assistance to avoid a severe physical withdrawal syndrome with associated risk of seizures).
[90] In 2013, almost 200,000 items of medication were prescribed (in a primary care setting or NHS hospital) for the treatment of AUD and dispensed in the community. The Net Ingredient Cost (NIC) of these prescription items in 2013 was 3.13 million.
In 2012, there were 6,490 alcohol-related deaths (HSCIC 2014). Taking into account alcohol related health disorders and disease, crime and antisocial behavior, accidents, loss of productivity in the workplace, and domestic problems, the Department of Health estimates that alcohol misuse is now costing around 20 billion a year in England alone (HM Government 2012).
[91] It is difficult to find agreement about outcomes of alcohol treatments because there are many different sorts of treatments, there are overlaps between treatments, and there are large differences between the sorts of patients, severity of disease, presentation of patients, and multiple confounding psychosocial factors. Nonetheless, the outcomes of current treatments are generally considered relatively poor.
[92] Licensed pharmacological options for treating MID include acamprosate, disulfiram, naltrexone, and nalmefene. The U.S. Food and Drug Administration (FDA) has approved only di sulfi ram, naltrexone, and acamprosate for the treatment of AUD, and the European Medicines Agency (EMA) has approved, in addition, nalmefene. Acamprosate, a glutamate antagonist, reduces NMDA activity associated with alcohol withdrawal and reduces relapse rates (Rosner et al. 2010). Disulfiram is used as an agent to deter relapse by preventing the elimination of acetaldehyde, which results in an unpleasant physical reaction if alcohol is used together (Krampe et al. 2006). Naltrexone is a competitive opioid antagonist which decreases cravings from alcohol (Soyka and Rosner 2008). Nalmefene is an opioid antagonist, similar to naltrexone but with a longer half-life and lower risk of toxicity than naltrexone. This can be given to patients while still drinking in order to reduce cravings (Paille and Martini 2014).
Benzodiazepines are commonly prescribed as a time-limited course as part of alcohol detoxification program (Lingford-Hughes et al. 2012).
[93] The efficacy of each of these above medications is only modest. Thus, there is still a high medical need to find more effective treatments for AUD. Numerous other pharmacological agents have been tested, although none have shown marked promise alone.
These include opioid antagonists (e.g., nalmefene), CBI antagonists (e.g., rimonabant), CRHI
receptor antagonists (e.g., verucerfont, pexacerfont), NK 1R antagonists (e.g., tradipitant), OTR
agonists (e.g., oxytocin), GABA agents (e.g., topiramate, baclofen, benzodiazepines), voltage-gated sodium channel inhibitors (e.g., oxacarbazepine, valproic acid, zonisamide), voltage-dependent calcium channel agonists (e.g., gabapentin, pregabalin), a7 nicotinic acetylcholine receptor agonists (e.g., varenicline), 5-HT 3 antagonists (e.g., ondansetron), 5-HT' A receptor partial agonists (e.g., aripiprazole), 5-HT2,6, receptor antagonists (e.g., quetiapine, olanzapine, mirtazapine), 5-HT reuptake inhibitors (e.g., trazodone), SERT inhibitors (e.g., duloxetine), ai adrenoreceptor antagonists (e.g., doxazosin, prazosin), glucocorticoid receptor antagonists (e.g., mifepristone), al adrenoreceptor agonists (e.g., guanfacine), AChE
inhibitors (e.g., citicoline), a dopamine D2 receptor antagonist (e.g., tiapride), and ct2 adrenoreceptor agonists (e.g., clonidine), among others.
[94] In terms of psychosocial interventions, the best approaches are those that foster good relationships and are delivered by the most experienced, highly trained therapists. However, the efficacy of current available treatments is far from satisfactory, with high rates of relapse.
A systematic review of 361 controlled studies of treatments for alcohol dependence in 2002 identified 46 possible interventions (Miller and Wilbourne 2002).
Interventions were ranked according to rates of abstinence achieved. The brief intervention approach ranked highest and motivational enhancement therapy, which is a counselling approach that helps individuals resolve their ambivalence about engaging in treatment and stopping their drug use, ranked second. The motivational enhancement approach aims to evoke rapid and internally motivated change, rather than guide the patient stepwise through the recovery process.
Pharmacotherapy with acamprosate and the opiate antagonist naltrexone ranked third and fourth respectively.
The lowest ranked approaches were designed to educate, confront, shock, or foster insight regarding the nature of alcoholism.
[95] A large three year follow-up study, project MATCH (Project MATCH Research Group 1997a), identified that 12-step facilitation (TSF), Motivational Enhancement Therapy (MET), and Cognitive-Behavioral Coping Skills Therapy (CBT) are in general equally effective. At three years the TSF clients 36% were abstinent, compared with 27% of MET and 24% of CBT
clients. A more recent review, the COMBINE Project (Anton et al. 2006), was designed to evaluate the efficacy of two relapse prevention medications in various combinations with behavioral treatment. Results indicated that both naltrexone and acamprosate showed only minor positive effects in relapse prevention, and only when used in conjunction with psychosocial interventions. A similar UK-based study, `UK ATT' (UK ATT
Research Team 2005), identified that Social Behavior and Network Therapy (SBNT) and MET
produced similar improvements in drinking and general functioning, with clients all reporting that total alcohol consumption had decreased by less than 50% at 12 months, with no significant differences in effectiveness between groups.
[96] In recent years, mindfulness techniques, originally derived from Vipassana meditation, which encourage awareness and acceptance of thoughts, feelings, and bodily sensations, have been explored as a potential approach for treating AUD (Marcus and Zgierska 2009).
Mindfulness promotes nonjudgmental acceptance of moment-to-moment thoughts.
This helps the person in recovery interrupt the tendency to respond to stress with alcohol use and not to react automatically to cravings (Shapiro 2006, Hsu et al. 2008). Although mindfulness shows promise as an adjunctive treatment to help individuals endure craving, evidence of its effectiveness has been limited, with many individuals needing further cognitive and behavioral skills to cope with high risk situations for relapse.
[97] 3,4-Methylenedioxymethamphetamine (MDMA) was first synthesized in 1912 by chemists at the pharmaceutical company Merck in Germany, as an intermediate in the synthesis of a compound, hydrastinine, that had been developed as a regulator of bleeding. In 1914 Merck was issued German Patent Nos DE274350 and DE279194 for that work MDMA at the time was referred to as "methylsafrylamin." MDMA however was not tested in humans until 1959.
Starting in 1977, following its introduction by Alexander Shulgin to psychiatrist Leo Zeff and subsequent widespread promotion by Zeff as an adjunct to psychotherapy, MDMA
became used therapeutically across the United States. Starting around 1979, non-clinical use of MDMA, now often called "ecstasy," began to grow, and came to the attention of U.S.
authorities, including the Drug Enforcement Administration (DEA).
[98] In 1985, because of this attention, MDMA was placed by emergency ruling in Schedule I of the Controlled Substances Act ___ the most restrictive class of drugs, reserved for substances that "have no currently accepted medical use," "a lack of accepted safety for use under medical supervision, and a high potential for abuse"¨and was similarly scheduled in other countries around the world. This resulted in no research being conducted into the potential therapeutic applications of MDMA between 1985 and 2000.
[99] However, starting in 1986, the Multidisciplinary Association for Psychedelic Studies (MAPS) began funding animal toxicity studies and human safety studies at Stanford University and Johns Hopkins University. In 1990, the first FDA approved, double-blind, placebo-controlled Phase I dose-response safety study of MDMA was published by Charles Grob, working with MAPS. In 2004, the first Phase 2 clinical trial of MDMA-assisted therapy for PTSD was approved by the FDA, sponsored by MAPS.
[100] By the time of this filing, MDMA has been included in over 5,000 academic articles and administered to over 1,100 human volunteers. Twelve clinical trials have been completed globally under sponsorship of MAPS, investigating MDMA-assisted psychotherapy for PTSD, anxiety related to advanced-stage illness, social anxiety in autistic adults, and an ongoing study of healthy subjects via an MDMA therapist training program.
[101] One Phase 1 study, "Open-Label Proof of Concept Feasibility Study to Explore the Safety, Tolerability and Potential Role of MDMA-Assisted Psychotherapy for the Treatment of Detoxified Patients With Alcohol Use Disorder," sponsored by Imperial College London, was the first to initiate an exploration that was expanded and built upon until disclosed herein (see also Sessa et al. 2021, which was still forthcoming at the time of the priority document of which this filing claims the benefit). The concept was that of the inventor hereof.
[102] Chemically, MDMA is a ring-substituted phenethylamine. It exerts its effects primarily through promoting raised levels of monoamine neurotransmitters in the brain, in particular serotonin, but also dopamine and noradrenaline. Increased activity at 5-HTIA
and 5-HTIB
receptors reduces feelings of depression and anxiety, reduces the amygdala fear response and increases levels of self-confidence (Brunner 1997). Furthermore, the effect of raised serotonin at 5-HT24 receptors provides alterations in the perceptions of meanings and facilitates new ways of thinking about old experiences (Nash et al. 1994). The effect of increased dopamine and noradrenaline is to raise levels of arousal and awareness, which increases sense of readiness and improves recall of state-dependent memories of stressful events (Cozzi 1999). Alongside this increase in arousal there is also a paradoxical increase in relaxation mediated by the effects of MDMA at alpha-2 receptors (Lavelle 1999). MDMA also has been shown to facilitate the release of oxytocin, which is the hormone associated with bonding and increased levels of empathy and closeness (Thompson 2007).
[103] MDMA produces elevated mood, increased sociability and feelings of closeness to others, slight alterations in perception that facilitate imagination and memory (Harris et al.
2002), greater compassion, feelings of sociability, closeness and increased empathy for others and themselves (Hysek et al. 2013) The effects of MDMA at reducing activity in the amygdala (Gamma et al. 2000) results in users experiencing a reduced fear response, and greater activity in the reward pathways in the Ventral Tegmental Area (VTA) in comparison to placebo, associated with less activity in the left amygdala after viewing angry faces, suggesting less reactivity to anger and improved prosocial behavior. Subjects receiving 1V1DMA
showed a better ability to detect positive facial expressions and a greater difficulty detecting negative facial expressions (Hysek et al. 2012a).
[104] These effects of MDMA give rise to its description as an "empathogen"
(meaning "generating a state of empathy," and independently suggested in 1983-84 by the psychologist and psychopharmacologist Ralph Metzner and the Purdue University professor of pharmacology and medicinal chemistry David Nichols) and -entactogen"
(subsequently coined by Nichols in 1986, and meaning "to touch within") (Holland 2001 at 182 n.2).
[105] Taken together, these changes in social cognition, interpersonal closeness, communication, and brain activity may influence the outcome of psychotherapeutic treatments for psychological disorders (Jerome et al. 2013). Nevertheless, the potential of MDMA to enhance the psychotherapeutic processes in the treatment of AUD was not before tested in a controlled clinical setting until the work of the inventor hereof.
[106] The use of MDMA-assisted psychotherapy has been explored as a treatment for post-traumatic stress disorder (PTSD) since the 1980s (Greer 1986, Sessa 2012) Long-term follow-up data from the first completed trial of MDMA-assisted psychotherapy for chronic, treatment-resistant PTSD found statistically and clinically-significant gains in symptom relief with no subjects reporting harm (Mithoefer et al. 2010; Mithoefer et al. 2013).
Another study in 12 subjects (Oehen 2013) showed significant self-reported improvement, with an effect size of 1.1 similar to Mithoefer et al. 2010. Findings from a recent randomized, double-blind, placebo-controlled, multi-site phase 3 clinical trial demonstrated that, compared to therapy with inactive placebo, MDMA-assisted therapy is highly efficacious in individuals with severe PTSD, and treatment is safe and well-tolerated, even in those with comorbidities (Mitchell 2021). Studies have also demonstrated potential for MDMA to address other difficult-to-treat mental health conditions, including obsessive compulsive disorder (OCD), phobias, eating disorders, depression, end-of-life anxiety, and social anxiety.
[107] How the primary actions of MDMA could treat AUD has not before been established or well-studied. In previously proposing a feasibility study, the inventor hereof postulated that MDMA-assisted psychotherapy may influence the outcome of psychotherapeutic treatments for AUD (Sessa 2017). However, evidence that would support an expectation that MDMA-assisted psychotherapy indeed would work for AUD did not then exist.
[108] Since the earliest days of psychedelic research in the 1950s, AUD has been a target for psychedelic-assisted psychotherapy ____ the theory being that an intense, drug-induced spiritual or mystical peak experience could be used as a method of inducing sobriety (Sessa, 2017).
LSD-assisted psychotherapy was explored with varying rates of success, and heterogeneity between studies. Early uncontrolled studies showed abstinence rates of 30-50%
(McLean et al., 1961; Kurland et al., 1967; Ditman & Bailey, 1967; Rydzynski et al., 1968).
[109] Drug-assisted psychotherapies with "classical" psychedelic compounds, such as LSD
and psilocybin, utilize the induced subjective mystical and spiritual effects of the psychedelic experience. Researchers have found that the depth of this experience is strongly associated with maintained abstinence from substance use (Bogenschutz 2015, Johnson et al.
2014, Kruspitsky 1997). In contrast, use of MDMA is generally not associated with a mystical experience. LSD
has been found to induce mystical experiences of significantly greater intensity relative to MDMA (Holze, 2019), as determined by the 5 Dimensions of Altered States of Consciousness scale (5D-ASC) (Dittrich, 1998) and the Mystical Experience Questionnaire (IVIEQ) (Barrett, 2015).
[110] Additionally, not all patients are able to tolerate the classical psychedelic experience, or even the idea of a treatment involving these experiences, and compliance is a critical part of addiction therapy. MDMA is generally more easily tolerated than the classical psychedelics, with less perceptually disturbing effects compared to LSD and psilocybin. But whether this in fact offers an alternative opportunity for enhanced psychotherapy was not before demonstrated.
Indeed, given that the depth of the mystical experience is strongly associated with maintained abstinence from substance use, it would have been logical to presume that MDMA-assisted therapy for AUD would have less success in treating patients than LSD- or psilocybin-assisted therapy (cf. McCulloch et al. 2021).
[111] Applicant discloses herein compositions of MDMA and their use in treating AUD, including both "dependent use" AUD and "harmful use" AUD. Using the compositions and methods of the invention, the psychotherapeutic processes involved in the treatment of AUD
are not only enhanced and intensified, but are improved in novel, unexpected, and surprising ways. As described below, the unique effects of MDMA, given in a psychotherapeutic context disclosed and using the compositions and methods of the invention, reduce avoidance of emotionally distressing thoughts, images, or memories while increasing empathy for the self and others. The compositions and methods of the invention herein also address symptoms of other conditions that are frequently comorbid with substance abuse, particularly those symptoms associated with a history of psychological trauma. Additionally, as taught herein, the compositions and methods of the invention can lead to reductions in alcohol cravings, problematic consumption, and rates of relapse.
[112] For these reasons and others, the disclosed invention represents a significant step toward solving the serious clinical, social, and personal problems caused by AUD. Among the various aspects of the present invention are compositions of MDMA and methods of their use in MDMA-assisted psychotherapy for the treatment of patients with AUD, including physically dependent alcohol use disorder ("dependent use" or "alcohol addiction") and non-physically dependent alcohol use disorder ("harmful use").
[113] MDMA-assisted psychotherapy, as described in the methods of the invention, may involve the careful screening of patients, such as through specific inclusion and exclusion criteria on which the success of treatment may depend. The therapeutic program involves both non-MDMA and MDMA-assisted sessions which are all delivered by trained healthcare professionals, in a safe and supportive setting. Visits involving MDMA dosing typically last 6-8 hours during which patients are carefully monitored.
[114] Using the compositions and methods of the invention, MDMA-assisted psychotherapy can be safely delivered, can be well tolerated, and can enhance and intensify the psychotherapeutic processes in the treatment of patients with AUD.
[115] MDMA, given in the psychotherapeutic context described by Applicant, can reduce avoidance of emotionally distressing thoughts, images, or memories of alcohol misuse, while increasing empathy for the self and others. It can also address symptoms of other conditions that are frequently comorbid with substance abuse, particularly those symptoms associated with a history of psychological trauma. Further, patients treated with the compositions and methods of the invention can reduce their alcohol consumption over the long term, resulting in reduced rates of relapse, and producing benefits not only to the individual patients treated, but to their friends, family, and society at large.
[116] As disclosed by Applicant and detailed below, MDMA-assisted psychotherapy is a superior method of treating patients with AUD, with numerous significant improvements over prior art methods. As shown below, MDMA-assisted psychotherapy has significant advantages over standard psychotherapy or other existing treatments for AUD, offering patients a potential new approach to a devastating, chronic, and relapsing condition.
[117] While the invention is described in terms of particular embodiments and applications, it is not intended that these descriptions in any way limit its scope to any such embodiments and applications, and it will be understood that many modifications, substitutions, changes, and variations in the described embodiments, applications, and details of the invention illustrated herein can be made by those skilled in the art without departing from the spirit of the invention, or the scope of the invention as recited in the appended claims.

A. Definitions and Terms
[118] The terminology used herein is for describing embodiments and is not intended to be limiting. As used herein, the singular forms "a," "an,- "and,- "the,- and "said" are intended to mean that there are one or more of the elements, and hence include plural referents, unless the content and context clearly dictate otherwise. Thus, for example, a reference to "an excipient"
may include a single such excipient, or a combination of two or more excipients.
[119] The terms "comprising," "including," "such as," and "having" are intended to be inclusive and not exclusive (i.e., there may be other elements in addition to the recited elements). Thus, the term "including" as used herein means, and is used interchangeably with, the phrase "including but not limited to." The term "or" is used herein to mean, and is used interchangeably with, the term "and/or," unless context clearly indicates otherwise.
[120] Unless otherwise indicated, all numbers expressing quantities used to describe and claim certain embodiments of the invention are to be understood as being modified in some instances by the term "about.- Accordingly, in some embodiments, the numerical parameters set forth in the description and claims are approximations that can vary depending upon the desired properties sought to be obtained by a particular embodiment. In some embodiments, "about" refers to plus or minus five percent ( 5%) of the recited unit of measure. The term -substantially," where it is applied to modify a parameter or characteristic herein, will be read in the context of the invention and in light of the knowledge in the art to provide certainty, e.g., by using a standard that is recognized in the art for measuring the meaning of "substantially"
as a term of degree, or by ascertaining the scope as would one of skill.
[121] In some embodiments, the numerical parameters should be construed in light of the number of reported significant digits and by applying ordinary rounding techniques.
Notwithstanding that the numerical ranges and parameters setting forth the broad scope of some embodiments of the invention are approximations, the numerical values set forth in the specific examples are reported as precisely as practicable. The numerical values presented in some embodiments of the invention may contain certain errors necessarily resulting from the standard deviation found in their respective testing measurements.
[122] Unless defined otherwise, all technical and scientific terms herein have the meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
Further definitions that may assist a reader to understand the disclosed embodiments follow.
[123] "Alcohol use disorder" or "AUD," in some embodiments, refers to the disorder as defined in the Diagnostic and Statistical Manual of Mental Disorders, 5th Edition (DSM-5).
Under DSM-5, anyone meeting any 2 of 11 listed criteria during the same 12-month period receives a diagnosis of AUD, the criteria being whether in the past year, a patient has: (1) Had times when you [i.e., the patient] ended up drinking more, or longer, than you intended? (2) More than once wanted to cut down or stop drinking, or tried to, but couldn't?
(3) Spent a lot of time drinking? Or being sick or getting over other aftereffects? (4) Wanted a drink so badly you couldn't think of anything else? (5) Found that drinking¨or being sick from drinking¨
often interfered with taking care of your home or family? Or caused job troubles? Or school problems? (6) Continued to drink even though it was causing trouble with your family or friends? (7) Given up or cut back on activities that were important or interesting to you, or gave you pleasure, in order to drink? (8) More than once gotten into situations while or after drinking that increased your chances of getting hurt (such as driving, swimming, using machinery, walking in a dangerous area, or having unsafe sex)? (9) Continued to drink even though it was making you feel depressed or anxious or adding to another health problem? Or after having had a memory blackout? (10) Had to drink much more than you once did to get the effect you want? Or found that your usual number of drinks had much less effect than before? (11) Found that when the effects of alcohol were wearing off, you had withdrawal symptoms, such as trouble sleeping, shakiness, restlessness, nausea, sweating, a racing heart, or a seizure? Or sensed things that were not there? The severity of AUD¨mild, moderate, or severe¨is based on the number of criteria met. For instance, in the 2015 National Survey on Drug Use and Health survey, wherein 43,561 individuals were polled, 11.8% met the criteria for alcohol use disorder-67% of which were "mild" cases (2-3 symptoms), 18.8% had a moderate disorder (4-5 symptoms), and 13.8% possessed a severe disorder (6 or more symptoms) (Witkiewitz, Litten, and Leggio, 2019).
[124] In some embodiments, AUD refers to a problematic pattern of alcohol use accompanied by clinically significant impairment or distress (Kranzler and Soyka, 2018).
In some embodiments, AUD refers to a disease characterized by a loss of control over alcohol drinking accompanied by changes in brain regions related to the execution of motivated behaviors and to the control of stress and emotionality (e.g., the midbrain, the limbic system, the prefrontal cortex, and the amygdal a) (Witkiewitz, Litten, and Leggio, 2019).
[125] Although herein the terms "alcohol use disorder" and "AUD" generally shall refer to the criteria in the DSM-5, or a patient with a diagnosis based thereon, it will be appreciated that the compositions and methods of the invention are equally applicable to patients having the equivalent underlying disorder, whether that disorder is diagnosed based on the criteria in DSM-5 or in DSM-IV (which described two distinct disorders¨"alcohol abuse- and "alcohol dependence"¨with specific criteria for each), whether the diagnosis is based on other clinically acceptable criteria (e.g., as "Alcohol Dependence Syndrome," see Edwards, G.
(1986), The Alcohol Dependence Syndrome: a concept as stimulus to enquiry.
Brit. J.
Addiction, 81:171-183), or whether the patient has not yet had a formal clinical diagnosis.
[126] "Alcohol dependence" refers to the more severe form of AUD involving physical alcohol dependence. Use of terms such as "physically dependent" alcoholism, "physically dependent" alcohol addiction, or "physically dependent" AUD, therefore will be understood to refer to the psychiatric disorder or condition in which an individual has become physically dependent upon, or physically addicted to, alcohol (e.g., Alcohol Dependence Syndrome).
Typically alcohol dependence is characterized by (often serious) withdrawal symptoms on cessation of alcohol, or drinking to avoid withdrawal symptoms, tolerance and the persistent desire to drink, and continuing drinking despite negative consequences (NICE
2013).
[127] "Harmful use of alcohol" or "harmful use" AUD refers to the definition in the WHO's International Classification of Diseases, 10th Revision (1CD-10 Classification of Mental and Behavioural Disorders) (1CD-10; WHO, 1992): "[A] pattern of psychoactive substance use that is causing damage to health. The damage may be physical (e.g., hepatitis) or mental (e.g., depressive episodes secondary to heavy alcohol intake). Harmful use commonly, but not invariably, has adverse social consequences; social consequences in themselves, however, are not sufficient to justify a diagnosis of harmful use."
[128] The distinction between the different severities of AUD can vary over time within a single patient's relationship with alcohol; however, one of ordinary skill will appreciate how to make an appropriate and reasonable clinical determination. Clinical signs and tools used to distinguish between those AUD patients with non-physical "harmful use" and those who are physically dependent on alcohol include whether or not there is evidence of physical dependence (including evidence of severe sweating, tremor, vomiting, nausea or confusion on cessation of drinking for =48 hours); whether or not there is evidence of a strong craving to drink alcohol in order to combat the symptoms of alcohol withdrawal; and whether one scores 15 or less (non-dependent, harmful use) or greater than 15 (dependent) on the Severity of Alcohol Dependence Questionnaire (SADQ). As the NICE Guidelines explain:
"Although alcohol dependence is defined in ICD-10 and DSM-IV in categorical terms for diagnostic and statistical purposes as being either present or absent, in reality dependence exists on a continuum of severity. Therefore, it is helpful from a clinical perspective to subdivide dependence into categories of mild, moderate, and severe. People with mild dependence (e.g., those scoring 15 or less on the Severity of Alcohol Dependence Questionnaire;
SADQ) usually do not need assisted alcohol withdrawal. People with moderate dependence (with a SADQ
score of between 15 and 30) usually need assisted alcohol withdrawal, which can typically be managed in a community setting unless there are other risks. People who are severely alcohol dependent (with a SADQ score of 30 or more) will need assisted alcohol withdrawal, typically in an inpatient or residential setting. In this guideline these definitions of severity are used to guide selection of appropriate interventions." In other embodiments, the categories of mild, moderate, and severe may be diagnosed based on the symptoms as defined by the DSM-5, wherein "mild" refers to two or three symptoms, "moderate" refers to four or five symptoms, and "severe" refers to six or more symptoms.
[129] AUD patients characterized as "harmful use," refer to those engaging in "harmful alcohol use," which is characterized by a pattern of use that causes damage to health, which may be physical or mental; and has adverse social consequences, including those affecting personal relationships, professional relationships, familial relationships, etc. (note, however, that adverse social consequences themselves do not justify a diagnosis of "harmful use"). Thus, in some embodiments, "harmful use" or "harmful alcohol use" refer to a condition where a patient scores between 0 and 15 on the SADQ. In some embodiments, "harmful use-or "harmful alcohol use" refer to a condition where a patient scores between 0 and 15 on the SADQ, and where the patient exhibits at least one of damage to health (e.g., physical, mental, or physical and mental health), and strained/damaged social relationships.
[130] The negative physiological effects of alcohol use are well-known and include damage to the gastrointestinal tract (Bishehsari, 2017; Egerer, 2005) and the liver (Ding, 2019). These effects may result in altered absorption and drug metabolism, depending on several factors, including whether alcohol consumption has resulted in changes to the structure and function of the gut and/or the liver. Alcohol use has also been shown to cause hypertension (Husain, 2014) and to compromise the cardiovascular system (Piano, 2017). Considering the hepatic metabolism of MDMA, including its conversion into hepatotoxic metabolites (de la Torre 2004; Carvalho, 2010), the safety, such as the type and severity of adverse events, and the utility of MDMA-assisted psychotherapy in subjects who consume alcohol, such as those diagnosed with AUD, may be unpredictable.
[131] "Alcohol use disorder patient" or "AUD patient" refers to a patient diagnosed with AUD (such as dependent use AUD or harmful use AUD), as defined herein. In some embodiments, the term -AUD patient" refers to a patient diagnosed with AUD who is in abstinence from alcohol, for example, for at least 1 day, such as 2 days or more, 3 days or more, 4 days or more, 5 days or more, 6 days or more, or 7 days or more. The phrase "AUD patient in abstinence from alcohol" refers to a patient diagnosed with AUD in abstinence from alcohol, i.e., having not consumed alcohol, for a given period of time, for example, for at least 1 day, such as 2 days or more, 3 days or more, 4 days or more, 5 days or more, 6 days or more, or 7 days or more. The term "AUD patient post-detox" refers to a patient diagnosed with AUD after having undergone a detoxification course, wherein a "detoxification course" or simply "detoxification" refers, in some embodiments, to the abrupt cessation of alcohol intake in individuals that suffer from alcohol dependence. In some embodiments, detoxification includes the administration of drugs to prevent symptoms of "withdrawal," which refers to a physiological response experienced by those having a physical dependence on alcohol, the symptoms of which include, but are not limited to, anxiety, shakiness, sweating, vomiting, fever, increased heart rate, seizures, hallucinations, and delirium tremens (National Clinical Guideline Centre, 2010). In some embodiments, a detoxification course may last for a given duration of time, for example, for at least 1 day, such as 2 days or more, 3 days or more, 4 days or more, 5 days or more, 6 days or more, or 7 days or more. In some embodiments detoxification or a detoxification course will be as specifically disclosed herein.
[132] "Binge drinking" refers to a pattern of drinking that brings a person's blood alcohol concentration (BAC) to at least .08 g/dl. A "binge drinker," i.e., one who binge drinks, is referred to as an "alcohol abuser" or "abuser of alcohol," wherein the act of binge drinking itself may be referred to as "alcohol abuse" or "abuse of alcohol." Abusers of alcohol may not drink on a consistent basis; for example, they may only drink once a week, but, when drinking, they may drink heavily, which may cause adverse health effects, such as, but not limited to, suffering from alcohol intoxication, unintentional injuries, such as car accidents, falls, burns, and alcohol poisoning; violence, including homicide, suicide, domestic violence, and sexual assault; sexually transmitted diseases; unintended pregnancy and poor pregnancy outcomes, including miscarriage and stillbirth; fetal alcohol spectrum disorders, sudden infant death syndrome, chronic diseases such as, but not limited to, high blood pressure, stroke heart disease, and liver disease; cancer, including but not limited to breast cancer, mouth cancer, throat cancer, esophogeal cancer, liver cancer, and colon cancer; memory and learning problems, and other behavioral addictions and substance use disorders (CDC, 2019).
[133] The frequency at which one binge drinks may determine whether or not the individual is considered a -heavy drinker" or -heavy alcohol user." -Heavy drinker"
refers to someone with a heavy alcohol use pattern The National Institute on Alcohol Abuse and Alcoholism (NIAAA) and the Substance Abuse and Mental Health Services Administration (SAMHSA) define "heavy alcohol use" as binge drinking on 5 or more days in the past month NIAAA
defines binge drinking as a pattern of drinking that brings BAC levels to at least 0.08 g/dL.
This typically occurs after 4 alcoholic drinks for women and 5 alcoholic drinks for men within a period of about 2 hours. SAMTISA defines "binge drinking" as 5 or more alcoholic drinks for males or 4 or more alcoholic drinks for females on the same occasion (i.e., at the same time or within a couple of hours of each other) on at least 1 day in the past month.
[134] "Alcohol," as used herein, for example in relation to "drinks,"
"alcoholic drinks," or "drinking," refers to ethyl alcohol (i.e., ethanol). "Alcohol craving" refers to a conscious desire or urge to consume alcohol. "Alcohol use" refers to alcohol consumption, and "alcohol consumption" refers to the standards known in the art, such as those of NIAAA
or WHO.
[135] "Reducing alcohol use" or "reduction of alcohol use" refers to reducing the amount or frequency of alcohol use, for example as assessed by urinalysis (e.g., by measuring metabolites of alcohol in urine, such as Ethyl Glucuronide (EtG)) or as assessed by using self-reported alcohol use with standardized tools like the Timeline Follow-Back self-report (Sobell & Sobell, 1996). In some embodiments, "reducing alcohol use" or "reduction of alcohol use" refers to a reduction in drinks per day, a reduction in drinks per drinking day, or a reduction in the frequency of drinking, such as a reduction in the percentage of drinking days or percentage of heavy drinking days. In some embodiments "reducing alcohol use" or the "reduction of alcohol use" refers to an increase in the time between consuming at least one drink of alcohol, such that less total units of alcohol are consumed within a given timeframe. A
"unit of alcohol"
refers to a standard drink, which contains about 14 g of pure alcohol (ethanol). For typical alcoholic beverages, this refers to 12 ounces of 5% alcohol beer, 5 ounces of 12% wine, and 1.5 ounces of 40% distilled spirits (NTH). "Drinking," "drinks," or "alcoholic drinks," is understood in the context of "standard drinks," such as spirits or blends that are intended for human consumption, wherein a "standard drink" equals 12 g (15.2 mL) ethanol. "Heavy drinking day"
refers to a day with a total alcohol consumption g (76.05 mL) of ethanol for men and k.40 g (50.7 mL) for women. In some embodiments, "reducing alcohol use" or the "reduction of alcohol use" refers to increasing the time to a "heavy drinking day," wherein "heavy drinking day" refers to a given 24-hour period in which the pattern of drinking meets that of "binge drinking," as defined herein.
[136] "Alcohol abstinence" or "in abstinence from alcohol" refers to the cessation of alcohol consumption for an extended period of time, for example, for at least 1 day, such as 2 days or more, 3 days or more, 4 days or more, 5 days or more, 6 days or more, or 7 days or more The term "promoting alcohol abstinence" or "promotion of alcohol abstinence"
refers to helping to maintain abstinence from alcohol use, in particular after at least 1 day of not drinking alcohol, for example maintaining abstinence from alcohol use for a period of days, including a period of a week or more, including more than one week, more than two weeks, more than three weeks, and a month or more, including more than one month, more than two months, more than three months, more than four months, more than five months, more than six months, more than seven months, more than eight months, more than nine months, more than 10 months, more than 11 months, and a year or more, including more than one year, more than two years, more than three years, more than four years, and more than five years, including values in between. In some embodiments, "promoting alcohol abstinence" or "promotion of alcohol abstinence" refers to helping to maintain abstinence from alcohol use, in particular after at least 1 day of not drinking alcohol, for example maintaining abstinence from alcohol use until at least the 3 month, 6 month, and/or 9 month follow-ups, as are disclosed within, or for longer than 9 months, or for longer than 1 year.
[137] "Relapse into alcohol use" or "relapse into alcohol consumption" refers to alcohol intake (i.e., the consumption of alcohol or the "taking" of alcohol) following a period of alcohol abstinence, for example following a period of alcohol abstinence of at least 1 day or more, such as 3 days, 1 week, 2 weeks, 3 weeks, 1 month, 3 months, 6 months, 9 months, 1 year, or greater than 1 year, and values in between. In some embodiments, "preventing relapse into alcohol use- or "preventing relapse into alcohol consumption- refers to the prevention of alcohol intake by an AUD patient after the patient has ceased the intake of alcohol for at least 24 hours. In some embodiments, "preventing relapse into alcohol use" or "preventing relapse into alcohol consumption" refers to the prevention of alcohol intake by an AUD patient after the patient has ceased the intake of alcohol for less than 24 hours. In some embodiments, "preventing relapse into alcohol use" or "preventing relapse into alcohol consumption" refers to the permanent cessation of alcohol intake. In some embodiments, wherein a subject is administered a composition of the invention, "preventing relapse into alcohol use" or "preventing relapse into alcohol consumption" refers to an increase in time prior to the resumption of alcohol intake as compared to the time prior to the resumption of alcohol intake by a subject who is not administered a compound of the invention. In some embodiments, the increase in time prior to the resumption can be, e.g., at least 1 day, such as 2 days or more, 3 days or more, 4 days or more, 5 days or more, 6 days or more, or a week or more, including more than one week, more than two weeks, more than three weeks, and a month or more, including more than one month, more than two months, more than three months, more than four months, more than five months, more than six months, more than seven months, more than eight months, more than nine months, more than 10 months, more than 11 months, and a year or more, including more than one year, more than two years, more than three years, more than four years, and more than five years, including values in between.
[138] "AUD associated with high risk drinking for acute problems"
refers to AUD associated with daily alcohol consumption __60 g/day (76.05 mL/day) of ethanol for men and g/day (50.7 mL/day) for women (International Guide for Monitoring Alcohol Consumption and Related Harm, WHO/MSD/MSB/00.4; WHO 2000, p. 51).
[139] "Impulsivity- refers to a predisposition toward rapid, unplanned reactions to internal or external stimuli with diminished regard to the negative consequences of these reactions to the impulsive individual or others" (Moeller et al. Am J Psych. 2001. 158: 1783-93).
[140] -Psychosocial or behavioral therapy" refers to, but is not limited to, such therapies other than the MDMA-assisted psychotherapy of the invention, e.g., 12-step facilitation therapy (e.g., NIA A A, Project MATCH Monograph Series Volume 1, NTH Publication No. 94-3722, (1995) reprinted 1999), cognitive behavioral therapy (e.g., as described in Arch.
Gen. Psychiatry 1999;
56:493-502), interpersonal therapy (e.g., as described in Psychol Addict Behav 2009; 23(1):
168-174), contingency management based therapy (e.g., as described in Psychol Addict Behav 2009; 23(1): 168-174; in J. Consul. Clin. Psychol. 2005; 73(2): 354-59;
or in Case Reports in Psychiatry, Vol. 2012, Article ID 731638), community reinforcement approach based therapy (e.g., as described in Drug Alcohol Depend 2004; 74:1-13), motivational interviewing based therapy (e.g., as described in J. Consul. Clin. Psychol.
2001; 69(5): 858-62), motivational enhancement based therapy (e.g., as described in Drug Alcohol Depend 2007, 91:97-101) or meditation based therapy, such as transcendental meditation based therapy (e.g., as described in Addiction 2004; 99(7):862-874 or J. Consul.
Clin. Psychol. 2000;
68(3): 515-52). -Standardized psychological treatment" or -standardized psychological support" refers to standardized sessions, which may be, e.g., once a week, twice a week, once every two weeks, once a month, and the like, in particular psychological treatment, support, or other counseling focused on alcohol consumption.
[141] "Mental health disorder," "mental illness," or "psychiatric disorder"
refer to a disease condition in a mammal, and preferably in a human, that generally involves negative changes in emotion, mood, thinking, and/or behavior. In some embodiments, "mental health disorder,"
"mental illness," or "psychiatric disorder" refer to a behavioral or mental pattern that causes distress or impairment of personal functioning (Bolton, 2008). Mental health disorders include post-traumatic stress disorder (PTSD), adjustment disorder, affective disorder, depression, atypical depression, postpartum depression, catatonic depression, a depressive disorder due to a medical condition, premenstrual dysphoric disorder, seasonal affective disorder, dysthymia, anxiety, phobia disorders, binge disorders, body dysmorphic disorder, a mood disorder related to another health condition, disruptive behavior disorders, eating disorders, impulse control disorders, obsessive compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), personality disorders, attachment disorders, dissociative disorders, and substance-related disorders, including alcohol or drug abuse or dependence disorders like AUD and SUDs (see Merck Manual of Diagnosis & Therapy, 20th Ed. (2018)).
[142] "Behavioral addictions" refer to non-substance addictions that are characterised by an inability to resist an urge to perform a behavior resulting in actions that are harmful to oneself or others (Grant, Schreiber, & Odlaug, 2013). Although behavioral addictions share some at least surface similarities with alcohol and substance abuse, they are separate and distinct categories and classifications. Examples of behavioral addictions include pathological gambling, kleptomania, pyromania, compulsive buying, compulsive sexual behaviour, Internet addiction, pornography, and binge eating disorder.
[143] "MDMA" refers to 3,4-methylenedioxymethamphetamine, i.e., 1-(1,3-benzodioxol- 5-y1)-N ¨methylpropan-2¨amine (IUPAC), whether as a racemate, as pure or substantially pure individual enantiomers, or as an enantiomerically enriched mixture in any amount greater than 0%, and whether in salt or freebase form, and includes polymorphs.
"MDMA" as used herein therefore will be understood to encompass all salt forms of MDMA, such as MDMA hydrochloride salt (3,4-MDMA HC1, available from, e.g., Cayman Chemical Co., Ann Arbor, MI, with purity k. 98%, as Item No. 13971, NSC 168383, CAS No. 64057-70-1). -MDMA" will include the hydrochloride salt in all hydrated crystalline forms, such as quarter-hydrate, hemihydrate, three-quarter hydrate, and monohydrate (see, e.g., Shulgin 1986, and references cited). It includes MDMA
HCI
"forms I-III" (see Nair 2021). It includes MDMA base, generally a colourless oil insoluble in water. It will be understood to include MDMA as a racemic mixture (i.e., S,R( )-3,4-methylenedioxymethamphetamine) (CAS 69610-10-2), as an enantiomerically enriched mixture (of whatever proportions), or as individual enantiomers (i.e., pure or substantially pure R-MDMA or S-MDMA). Both enantiomers function as monoamine releasers (Hiramatsu and Cho, 1990; Johnson et al. 1986; Setola et al. 2003), with S-MDMA being a more potent dopamine releaser, and preclinical evidence suggesting that R-MDMA may provide an improved therapeutic index, maintaining the therapeutic effects of ( )-MDMA
with a reduced side effect profile (see, e.g., Anderson et al. 1978; Curry 2018; Pitts 2018).
"MDMA" thus may refer to racemic MDMA or to enantiomerically enriched compositions (e.g., to balance the individual therapeutic effects of S-MDMA and R-MDMA, or to minimize certain negative effects, such as the suggested greater neurotoxicity of the S-(+) enantiomer) or compositions of individual MDMA enantiomers. It also includes salt forms of individual enantiomers, e.g., S-MDMA HC1 (CAS 69558-32-3) and R-MDMA HC1 (CAS 69558-31-2), or mixtures in any portions thereof. In enantiomeric or enantiomerically enriched embodiments, the dosage may be adjusted accordingly based on the known differences in potency, and may be done so based on the teachings herein in combination with the general knowledge in the art.
Accordingly, all reference to "MDMA" (unless stated or necessarily implied otherwise, or where needed to provide technical effect) will be understood to include single enantiomers, enriched mixtures, and racemic mixtures, as well as all pharmaceutically acceptable salts, hydrates, solvates, co-crystals, polymorphs, prodrugs (e.g., as defined below), or metabolites thereof (e.g., 3,4-methylenedioxy- amphetamine (1VIDA), 4-hydroxy-3-methoxymethamphetamine (I-EVIMA), 4-hydroxy-3- methoxyamphetamine (HMA), 3,4-dihydroxyamphetamine (HHA), and 3,4-dihydroxy- methamphetamine (HEIMA)), and combinations thereof
[144] As used herein, the terms "subject," "user," "patient," and "individual"
are used interchangeably, and refer to any mammal although preferably a human. In some embodiments, the terms -subject," -user," -patient," and -individual" refer to any mammal, although preferably a human, who has an indication for which a compound, composition, or method described herein may be efficacious, or who otherwise may benefit by the invention. In general, all of the compounds, compositions, and methods of the invention will be appreciated to work for all individuals, although individual variation is to be expected, and will be understood. In some embodiments, as used herein, the terms "subject," "user," "patient," and "individual"
includes one who has a mental health disorder, or a condition related to a mental health disorder for which similar treatment may be efficacious; in embodiments herein, unless otherwise specified, a "mental health disorder" shall refer to an alcohol use disorder.
In embodiments herein, unless otherwise specified, an "alcohol use disorder" shall refer to both "harmful use"
and "dependent use" AUD. When a "subject," "user," "patient," or "individual"
is participating in a course of therapy as described below, the term "participant" also may be used. These terms also shall refer to patients in need of treatment for such a disorder, persons predisposed to such a disorder, and subjects whether or not diagnosed with such a disorder.
Moreover, these terms shall likewise refer to persons who have received treatment or therapy for a mental health disorder, are currently receiving therapy or treatment for a mental health disorder, or who may receive therapy or treatment for such a disorder in the future. In some embodiments, the disclosed methods al so can be used to improve mental health and improve psychological functioning in non-disease states, i.e., in an individual without a diagnosed mental health disorder, or specific symptoms thereof.
[145] In general, all of the compositions and methods of the invention will be appreciated to work for all individuals, although individual variation is to be expected, and will be understood.
Where there is variation between individuals, modification to the compositions and methods will be understood based on the teachings herein in combination with the general knowledge of the art. In some instances, certain personalized approaches (i.e., "personalized" or "precision" medicine) may be utilized, based on individual characteristics, including drug metabolism (e.g., CYP2D6 or CYP3A4) or individual genetic variation. The term "genetic variation" refers to a change in a gene sequence relative to a reference sequence (e.g., a commonly-found and/or wild-type sequence). Genetic variation may be recombination events or mutations such as substitution/deletion/insertion events like point and splice site mutations.
In one embodiment, the genetic variation is a genetic variation in metabotropic glutamate receptor type 5 (mGluR5), which has been implicated in mood and anxiety symptoms in humans, as well as playing a direct role in the pathogenesis of AUD, showing involvement in the development of behavioral sensitization towards ethanol in animal models.
In another embodiment, the genetic variation is one or more single nucleotide polymorphisms (SNPs) in the FKBP5 gene that are associated with elevated levels of FKBP51 protein relative to persons lacking such SNPs. The FKBP5 gene has been implicated in responses to stress and trauma, and such SNPs are correlated with susceptibility to certain depression, PTSD, and anxiety disorders (Yehuda 2016, Bierer 2020).
[146] In one embodiment, the genetic variation is a genetic variation in one or more cytochrome P450 (CYP or CYP450) enzymes that affects drug metabolism, including metabolism of a composition of the invention, and including CYP1A2, CYP2C9, CYP2D6, CYP2C19, CYP3A4 and CYP3A5. In other embodiments, CYP enzymes include CYP1A1, CYP1B1, CYP2A6, CYP2A13, CYP2B6, CYP2C8, CYP2C9, CYP2C18, CYP2E1, CYP2G1, CYP2J2, CYP2R1, CYP2S1, CYP3A5P1, CYP3A5P2, CYP3A7, CYP4A11, CYP4B1, CYP4F2, CYP4F3, CYP4F8, CYP4F11, CYP4F12, CYP4X1, CYP4Z1, CYP5A1, CYP7A1, CYP7B1, CYP8A1, CYP8B1, CYP11A1, CYP11B1, CYP11B2, CYP17, CYP19, CYP21, CYP24, CYP26A1, CYP26B1, CYP27A1, CYP27B1, CYP39, CYP46, and CYP51.
[147] In some embodiments, a composition of the invention is taken together with a compound that is metabolized by the same CYP enzyme(s) as the composition of the invention, so as to permit a lower dose to be taken, increase the effective bioavailability of one or both, or otherwise affect drug metabolism or pharmacokinetics In some embodiments, the dose of a composition of the invention is adjusted when administered to a subject known to be a "poor metabolizer" of the active agent in composition (e.g., having a genetic variation in CYP2D6, known to be the major metabolizer of the methylenedioxy moiety of MDMA).
[148] In some embodiments, a genetic variation (or a genetic marker signifying a variation) is an exclusion criteria for the administration of a compound of the invention. In one embodiment, the genetic variation is a genetic variation in metabotropic glutamate receptor type 5 (mGluR5), which has been implicated in mood and anxiety symptoms in humans. In another embodiment, the genetic variation is one or more single nucleotide polymorphisms (SNPs) in the FKBP5 gene that are associated with elevated levels of FKBP51 protein relative to persons lacking such SNPs. The FKBP5 gene has been implicated in responses to stress and trauma, and such SNPs are correlated with susceptibility to certain depression and anxiety disorders and PTSD. In one embodiment, the genetic variation is a genetic variation such as a SNP in a membrane transporter, such as SERT, DAT, NET, or VMAT. In one embodiment, the mammal being treated has altered epigenetic regulation of a gene the expression of which is associated with a mental health condition or susceptibility to a mental health treatment, such as the SIGMAR1 gene for the non-opioid sigma-1 receptor.
[149] In some embodiments, any modification to the compositions and methods of the invention are determined by the age of the patient, the weight of the patient, whether the patient has any comorbidities (i.e., any other medical conditions simultaneously present within a patient), other medications the patient is taking (routinely or presently), any patient-specific aspects that could affect the way in which the compositions of the invention interact with the patient, such as, but not limited to, variations in metabolism and variations in patient response, and any other such natural variations that would be apparent to one of skill.
[150] "Therapist" refers to a person who treats a patient using the compositions and methods of the invention, whether that person is a psychiatrist, clinical psychologist, clinical therapist, psychotherapist, or other trained counselor, facilitator, or guide, and who may or may not be a trained addiction specialist or counselor, substance abuse specialist or counselor, or alcohol abuse specialist or counselor. Generally, the therapist will be certified in the use of the treatment manual for the MDMA-assisted psychotherapy administered, and will have completed the appropriate training in delivering that form of MDMA-assisted psychotherapy.
This will enable a therapist to respond in a safe and supportive manner during the non-MDMA
and MDMA sessions. Studies with MDMA-assisted psychotherapy have shown that most psychiatric emergencies, e.g., acute anxiety associated with the emergence of challenging emotional material during the therapeutic session, can be dealt with through "talking down" an anxious patient, with no necessary requirement for psychiatric medication.
However, within the definition of "therapist" are psychiatrists licensed to manage psychiatric emergencies by administering "rescue medications- such as short-acting benzodiazepines if clinically indicated.
[151] -Treating" or -treatment" of a disorder includes: (i) inhibiting the disorder, i.e., arresting or reducing the development or progression of the disorder or its clinical symptoms;
or (ii) relieving the disorder, i.e., causing regression of the disorder or its clinical symptoms.

Inhibiting the disorder, for example, would include prophylaxis. Hence, one of skill will understand that a therapeutic amount necessary to effect treatment for purposes of this invention will, for example, be an amount that provides for objective indicia of improvement in patients having clinically-diagnosable symptoms. Generally, "objective indicia" will be those recognized by clinicians who diagnose and treat patients having the condition, and therefore readily appreciated in the art. In some embodiments, -objective indicia" may include, but are not limited to, an improvement in weight, body temperature, heart rate (FIR), respiratory rate, blood oxygenation, blood pressure (BP) and its variables, including, but not limited to:
systolic (SBP), diastolic (DBP), mean arterial (MAP), and pulse (PP);
continuous non-invasive beat-by-beat blood pressure (CNIBP); measurements from an electrocardiogram (ECG), including, but not limited to, RR interval or its variability, QT interval or its variability, heart rate variability (HRV) (or measured by devices other than an ECG); hemodynamic response (HR), and levels of glucose, cortisol, serotonin, dopamine, cholesterol;
electroencephalography (EEG) measures such as quantitative EEG (qEEG); electrocochleogram (ECochG), electromyography (EMG), electrooculography (EOG), magnetoencephalography (MEG);
el ectrocorticography (ECoG); magnetic resonance imaging (MRI); functional MRI
(fMRI);
computed tomography (CT); positron emission tomography (PET); nuclear magnetic resonance (NIVIR) spectroscopy or magnetic resonance spectroscopy (TVISR);
single-photon emission computed tomography (SPECT); near infrared spectroscopy (NIRS); event-related optical signal (EROS); computed axial tomography; diffuse optical imaging (DOT); cranial ultrasound, or functional ultrasound imaging (fUS) (together, "EEG measures"), brain derived neurotrophic factor (BDNF); genetic markers including relating to CYP enzymes or drug metabolism; genetic variation in mGluR5 or FKBP5; and numerous more, as will be readily appreciated by those of skill. In some embodiments, the objective indicia of improvement may be an improvement in one's psychological state, wherein "psychological state-may refer to the condition or state of one's mind including, but limited to, its physiological and subjective characteristics. In some embodiments, psychological state is synonymous with the terms or phrases "mental condition," "mental state," "psychological condition," "neural status,"
"cognitive state," "state of mind," "frame of mind," "emotional state," etc.
Physiologically, in the context of the invention, psychological state broadly refers to the condition of the nervous system and its components, including, but not limited to, the brain and its subunits, non-limiting examples of which include the frontal lobe, temporal lobe, brain stem, cerebellum, occipital lobe, parietal lobe, and cerebral cortex. Regardless, the effect, or improvement, may be prophylactic in terms of completely or partially preventing a disorder or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disorder, an adverse effect attributable to the disorder, and/or a comorbidity simultaneously present with the disorder. A
"comorbidity- present with AUD includes psychiatric disorders such as antisocial personality disorder, borderline personality disorder, depression, anxiety, schizophrenia, attention deficit hyperactivity disorder, bipolar disorder, obsessive compulsive disorder, binge eating disorder, and PT SD.
[152] Moreover, "treatment" as used herein covers any treatment of a disorder in a mammal, and preferably in a human, and includes: (a) preventing a disorder from occurring in a subject who may be predisposed to the disorder but has not yet been diagnosed with it;
(b) inhibiting a disorder, i.e., arresting its development; (c) relieving a disorder, i.e., causing regression thereof, (d) protection from or relief of a symptom or pathology caused by or related to a disorder; (e) reduction, decrease, inhibition, amelioration, or prevention of onset, severity, duration, progression, frequency or probability of one or more symptoms or pathologies associated with a disorder; and (f) prevention or inhibition of a worsening or progression of symptoms or pathologies associated with a disorder or comorbid with a disorder. Other such measurements, benefits, and surrogate or clinical endpoints, alone or in combination, would be understood to one of skill based on the teachings herein and general knowledge in the art.
[153] Moreover, any of the disclosed methods can be used in combination with one or more therapeutically beneficial activities, where such participation follows or is in conjunction with the admini strati on of the compositi on, including breathing exercises, rn editati on and concentration practices, focusing on an object or mantra, listening to music, physical exercise, stretching or bodywork, journaling, grounding techniques, positive self-talk, or engaging with a pet or animal, and it should be understood that such participation can occur with or without the participation or guidance of a therapist or other professional.
[154] An exemplary treatment protocol is outlined in FIG. 15, and is further detailed as follows. In this exemplary embodiment, a patient will first be assessed to see if they meet the inclusion protocol, which contains criteria outlined in TABLE 1 (1501). If not (1503), the patient is excluded from the treatment protocol (1504). If all inclusion criteria are met (1502), however, the patient is further assessed to determine if any of the exclusion criteria (also listed in TABLE 1) are met (1505). Unlike inclusion criteria, in which all must be met, only one exclusion criteria need be present (1506) to exclude the patient from participating in the treatment protocol or method of the invention (1508). If no exclusion criteria are found to be met (1507), however, the patient may participate in the treatment protocol/method of the invention (1509). While participating in a treatment protocol or method of the invention, the patient will further be assessed for withdrawal criteria (1510), also listed in TABLE 1, as with exclusion criteria, if one is met (1511), the patient is withdrawn from the treatment protocol or method of the invention (1513). So long as no withdrawal criteria are met (1512), the patient may continue with the treatment protocol or method of the invention, and it thus be determined as to whether or not the patient satisfies the requirements of the particular treatment protocol or method of the invention (1514). If so (1515), the patient thereby completes the treatment protocol (1518). However, if the patient does not satisfy the requirements of the particular treatment protocol/method of the invention (1516), the patient is withdrawn from the treatment protocol, or method of the invention (1517).
[155] For example, patients undergoing MDMA-assisted psychotherapy may be withdrawn prior to or during a course of therapy if certain withdrawal criteria were to occur. However, after patients have completed a course of therapy (e.g., an exemplary 8-week course or 13-week course of MDMA-assisted psychotherapy as in embodiments below, including those of Examples 4 and 8) they generally are not withdrawn based on these criteria.
Patients are always free to withdraw at any time from treatment without giving reasons and without prejudicing further treatment.
[156] Examples of "inclusion criteria," "exclusion criteria," and "withdrawal criteria" within the meaning of the invention include those in TABLE 1 below. It will be readily appreciated that such criteria are exemplary, optional, and non-limiting, unless stated or necessarily implied otherwise, or where needed in embodiments to provide technical effect.
TABLE 1: Exemplary Inclusion, Exclusion, and Withdrawal Criteria Exemplary Inclusion Criteria 1 Informed consent 2 Primary diagnosis (e.g., as defined by DSM-5) of alcohol use disorder (including, e.g., dependent use or harmful use AUD) 3 Successful alcohol detoxification (no longer consuming any alcoholic substances) (for "dependent use" patients); successful completion of a preparatory group therapy course (for "harmful use" patients; for some "dependent use" patients) 4 Between 18 and 65 years old Be able to identify in advance a supportive friend, family member, or significant other:
= who could meet with the clinician and receive information about the treatment at one of the preparation sessions prior to the patient's first MDMA session = who could accompany the patient to study visits if required = who could act as a "locator" and be contacted by the therapy team in order to remind the patient about follow-up appointments or collect outcome data (such as drinking behavior) in the event that the patient themselves cannot be contacted 6 Proficient in speaking and reading the language in which the therapy is provided 7 Agree to comply with requirements of therapy protocol Exemplary Exclusion Criteria 1 Lacking capacity 2 Personal history of, or a current, primary psychotic disorder, bipolar affective disorder type 1 or personality disorder 3 Present a serious suicide risk, e.g., as determined using the clinical judgment of one or more qualified mental health professionals within a therapy team. Risk may be determined using information from the Columbia-Suicide Severity Risk Scale (C-SSRS) which allows classification of severity of suicidal ideation and behavior. The C-SSRS scale classifies severe risk as (a) current suicidal ideation with intent and/or plan, and (b) suicidal behavior in the last 3 months. A clinical judgment regarding the level of risk and subsequent decisions regarding eligibility and care could use a combination of the information provided by the C-SSRS, the patient's history of previous risk behaviors, any presenting mental health difficulties, and environmental and clinical factors. A final decision may include a discussion with the qualified mental health professionals within the therapy team.
4 Relevant abnormal clinical findings at the screening visit.
These include:
= History of cardiac disease or stroke = History of severe liver disease, as evidenced by abnormal liver function test results, particularly reduction in albumin (normal > to 3.5 gm/di) = History of epilepsy = History of Malignant Hyperthermia (Central Core Disease) = Current untreated/unstable hypertension 5 Regular user of recreational MDMA or "ecstasy," e.g., more than five times in the last five years or at least twice in the 6 months prior to the start of the study 6 Currently taking or unwilling/unable to stop any medications inhibiting CYP2D6, and the following medications: monoamine oxidase inhibitors, Ritonavir, paroxetine, fluoxetine, citalopram, regular benzodiazepines or any other medications likely to interact with MDMA; to be stopped during course of MDMA-assisted therapy only (e.g., during exemplary 8 or 13 week courses below) 7 Regular use of or particularly dependence on other drugs such as benzodiazepines, synthetic cannabinoids, ***e, or heroin 8 For females of childbearing age and potential: must not be pregnant and/or breast-feeding, until the end of the treatment phase 9 Patients that might face additional risks from immunosuppression (for example patients with immunological diseases, patients with active infection or history of infections within four weeks of MDMA administration, etc.) Exemplary Withdrawal Criteria 1 Development of concurrent physical illness or deterioration of mental health warranting additional interventions and support that necessitates removal from therapy 2 The patient becomes pregnant 3 Start of medications that may interact with MDMA (see exclusion criteria above) 4 Levels of drinking that interfere with the patient's ability to engage with the treatment. If a patient has an isolated lapse to drinking, participation may be continued once stable enough to engage in the treatment.
If a patient is using illicit substances that are considered to interact adversely with the MDMA sessions or other aspects of the psychotherapy course
[157] In some embodiments, patients will be required to taper and stop any medications with a known or suspected risk of drug interaction with MDMA. "Drug interaction"
refers to a case wherein two or more drugs react with one another in the body and, as a result, either potentiate the effects of one or both of the drugs (i.e., increase the strength with which the drugs act in the body such that their effects are greater than would be expected on their own), or cause unexpected, and often adverse, side effects. In some embodiments, drug interactions also refer to drug and food, or drug and beverage interactions, wherein the drug administered to the patient interacts with a given food or beverage (namely, the compounds within the food or beverage), to cause an unexpected, often adverse reaction. In some embodiments, drug interactions also refer to drug and condition interactions, wherein administration of a drug may worsen a diagnosed, or undiagnosed medical condition.
[158] Generally, medication will only be tapered and stopped if deemed appropriate based on clinical decision and discussion with the therapy team. A drug or substance being "tapered,"
or "tapering" a drug or substance, refers to the process of slowly decreasing the amount of the drug or substance being taken over a given period of time. Meaning, the subject is administered smaller, and smaller amounts of the drug or substance to avoid or limit withdrawal symptoms, which would likely occur if the subject were to quit taking the drug or substance "cold turkey,"
(i.e., without tapering). As apparent to one of skill, tapering may be completed through direct tapering, which involves slowly reducing the amount of the drug or substance taken over a given amount of time (which is dependent upon the drug or substance taken, the individual, the length of use and other such metrics that would be known to those of skill);
substitution tapering, wherein the drug or substance is replaced with a drug or substance that, in some embodiments, causes a similar physiological response, but has a longer half-life, so tapering is more effective; and titration tapering, wherein the drug or substance is dissolved in a specific amount of a fluid, generally but not necessarily water, so that the "strength"
of each dose, or amount of the drug or substance in each dose administered to the patient, may be less than the previous dose (Coalition Recovery, 2019).
[159] Regardless of the tapering method, the washout period of medications to be stopped will be dependent on the half-life of that particular medication. In some embodiments, a sufficient washout period of 2 weeks (5 weeks for fluoxetine) must be allowed prior to the first MDMA session and, in such embodiments, medications should not be restarted until completion of the final MDMA session unless clinically required. Exemplary psychotropic medication that generally will be excluded include: any drugs inhibiting the liver enzyme CYP2D6, antidepressants of the serotonin and/or noradrenaline reuptake inhibitor type (SSRUSNRI), Mirtazapine, Ritonavir and regular use of benzodiazepines. These psychotropic medications may either elevate the risk of serotonin syndrome when combined with MDMA, or may reduce the therapeutic effects of MDMA (Hysek 2012b). By contrast, exemplary medications that generally will be permitted include: pregabalin, gabapentin, occasional benzodiazepines and Z-hypnotics (e.g., zolpidem, zopiclone, zaleplon, or eszopiclone).
Commonly used medications during and after the detoxification processes that generally will be permitted include baclofen, acamprosate, naltrexone, and disulfiram.
Evidence suggests there is an apparent decrease in CYP3A4 activity after a single dose of MDMA
of 1.5mg/kg (Yubero-Lahoz 2011), and in some embodiments certain CYP3A4 medications therefore may be contraindicated. Adjustments may also be made for patients who are considered "slow" or "fast" metabolizers, e.g, for CYP2D6 and/or CYP3A4 (see, e.g., Zanger 2013).
[160] "Assessment" refers to any means or method used with a patient, whether before, during, after, or unrelated in time to a specific treatment protocol, to measure, estimate, or evaluate a nature, ability, symptom, disorder, or other characteristic of the patient, whether qualitatively or quantitatively, and whether performed by the therapist or other clinician (e.g., an interview), by the patient his or herself (e.g., a self-reported questionnaire), by a third-party human or by a medical or other device (e.g., a medical sensor or biosensor, a watch or fitness tracker), and whether graded by a human decision-maker or an artificial intelligence (Al), using machine learning, or with a computer algorithm. Non-limiting examples of assessments include those in TABLE 2 below.
TABLE 2: Exemplary Patient Assessments 1 The Severity of Alcohol Dependence Questionnaire (SADQ) (Stockwell et al. 1983).
This is a 20 item clinical screening tool designed to measure the presence and level of alcohol dependence, taking approximately five minutes. This scale is designed to assess people that are currently drinking and therefore generally ("generally," here and elsewhere, means "in a preferred embodiment," but does not mean "necessarily") will only be administered once, at screening before patients undergo alcohol detoxification.
2 The Mini International Neuropsychiatric Interview 5 (MINI 5) (Sheehan et al. 1998) to screen for comorbid psychiatric disorders. This generally will be completed in full at screening, once, taking approximately 20 minutes.
3 The Columbia Suicide Severity Rating Scale (C-SSRS) (Mundt, JC
et al. 2013), to screen for acute and recent suicide and self-harm thoughts and behaviors, taking approximately five minutes to complete. Generally to be completed at screening, at baseline after detox, before each of the two MDMA-assisted sessions, the days after each MDMA session and in the exemplary embodiments, at 3, 6 and 9-month follow-up.
4 The Alcohol Dependence Syndrome section of the SCI-5-CT
(Clinical Trials Version) (First et al. 2015), this semi-structured interview generally will be conducted by a trained member of the therapy team and completed once, at screening, taking approximately ten minutes.
The Patient Health Questionnaire (PHQ-9) (Kroenke et al. 2001). A brief self-administered screening questionnaire for depressive symptoms. It will be administered at screening, baseline, the final therapy session and at 3, 6 and 9 months' follow-up (i.e., here and elsewhere, if follow-up is performed), taking approximately five minutes.
6 Generalized Anxiety Disorder 7 (GAD-7) (Spitzer et al. 2006) is a self-reported questionnaire for screening and severity measuring of generalized anxiety disorder (GAD). It generally will be administered at screening, baseline, the final therapy session and at 3, 6 and 9 months' follow-up, taking approximately five minutes.
7 Short Inventory of Problems for Alcohol (SIP) (Kiluk et al.
2013) SIP for alcohol is a 15-item instrument that assesses the self-attributed consequences of drinking taking approximately 5 minutes. This generally will be completed at screening, baseline, the final therapy session and at 3, 6 and 9 months' follow-up.
8 The Alcohol Timeline Follow Back (TLFB) (Sobell 2001) is a method of assessing daily drinking and recreational drug use over time. Patients generally will be given the TLFB
diary after screening, data will be collected at each therapy session. After completion of therapy, a researcher generally will call on a monthly basis to collect data throughout the study. Further discussion of the diary generally will take place formally at 3, 6 and 9 months follow up.
9 The Clinical Institute Withdrawal Assessment for Alcohol¨Revised Version (CIWA¨Ar) (Saitz et al. 1994). This is a ten-item scale used in the assessment and management of alcohol withdrawal. Each item on the scale correlates to the severity of alcohol withdrawal. This generally will be administered at baseline (after detox) and before each MDMA-assisted session, taking approximately 3 minutes.
The Penn Alcohol Craving Scale (PACS) (Flannery et al. 1999) is used to assess frequency, intensity and duration of thoughts about drinking, completed at the screening and baseline assessment and at 3, 6 and 9 months' follow-up.
11 Obsessive Compulsive Drinking Scale (OCDS) (Anton 1995) is used to measure obsessive and compulsive thoughts in relation to alcohol. Completed at the screening and baseline assessment and at 3, 6 and 9 months' follow-up.
12 Pittsburgh Sleep Quality Index (PSQI) (Buysse 1989) is used to assess the level of sleep disturbance, completed at screening, the baseline assessment and at 3, 6 and 9 months' follow-up.
13 Interpersonal reactivity Index (IRI) (Davis 1980) comprises 28 items answered on a 5 point scale. This scale measures different aspects of empathy and provides different sub scal es relating to these. This generally will be administered at baseline, the final therapy session, 3, 6 and 9 months' follow-up to explore if these core aspects of empathy are modified by MDMA Assisted therapy.
14 The Short Form (36) Health Survey (SF-36) is a gold standard patient-reported measure of quality of life that can be completed at baseline, 3, 6 and 9 months.
The Self-Compassion Scale (SCS) (Neff 2003) Comprises 26 items answered on a 5 point scale. This scale measures core aspects of self-compassion including components of mindfulness. This generally will be administered at baseline, the final therapy session, 3, 6 and 9 months' follow-up.
16 The Trauma History Questionnaire (THQ) (Green 1996) is a self-report measure that examines experiences with potentially traumatic events using a yes/no format.
For each event endorsed, respondents are asked to provide the frequency of the event as well as their age at the time of the event. It generally will be administered once, at baseline
[161] An assessment may be computer-assisted, and other computer-assisted assessments may be performed besides the assessments above. The term "computer-assisted"
in "computer-assisted assessment" means an assessment comprising the use of electronic tools such as online tools, smartphones, wireless devices, or health apps (in some such examples, also known as "digital phenotyping"). Computer-assisted assessment may include use of an electronic psychiatric notes system, where relevant clinical information is recorded for the duration of the therapy by a therapist interacting face-to-face with a patient, and will also include the use of computer systems where the therapist and patient interact virtually (either synchronously or asynchronously), as well as where a patient only interacts with a computer ("computer" broadly meaning any electronic tool suitable for such purposes, including desktop, laptop, and notebook computers; tablets, smartphones, and other mobile devices; watches, fitness trackers, and personal electronic devices; and the like). "Computer-assisted" may refer to one or more aspects of a psychosocial, behavioral, or MDMA-assisted therapy.
[162] "Therapeutic effect" or "therapeutic efficacy," as used herein, means the response(s) in a mammal, and preferably a human, after treatment that are judged to be desirable and beneficial. Hence, depending on the disorder to be treated, or improvement in physiological or psychological functioning sought, and depending on the particular constituent(s) in the compositions of the invention under consideration, those responses shall differ, but would be readily understood by those of ordinary skill. For AUD, including for both "harmful use" AUD
and "dependent use" AUD, those responses will be understood based on the teachings herein and the general knowledge in the art.
[163] Measures of therapeutic effect includes any outcome measure, endpoint, effect measure, or measure of effect within clinical or medical practice or research which is used to assess the effect, both positive and negative, of an intervention or treatment, whether patient-reported, gathered through laboratory tests such as blood work, urine samples etc., or through medical examination. In some embodiments, therapeutic effect may include the collection of objective measures, including but not limited to weight, body temperature, heart rate, respiratory rate, blood oxygenation, BP and its variables, including SBP, SBP, MAP, and PP;
CNIBP; ECG measurements, including, but not limited to, RR interval or its variability, QT
interval or its variability, HRV (including measured by devices other than an ECG);
hemodynamic response, and levels of glucose, cortisol, serotonin, dopamine, cholesterol; EEG
measures; BDNF; genetic markers including relating to CYP enzymes or drug metabolism;
genetic variation in mGluR5 or FKBP5; and numerous more, as will be readily appreciated by those of skill.
[164] Measures of therapeutic effect for purposes of the invention will be understood to include, as non-limiting examples: (1) Weekly units of alcohol consumed post-detox (i.e., counting from the last day of an alcohol detoxification course), or post "baseline" as defined herein, wherein a unit of alcohol refers to a standard drink that contains about 14 g of pure alcohol (ethanol), or other measure of relapse status; (2) Quality of life, including subjective sleep; (3) Psychosocial functioning; (4) Prescribed medication use; (5) Number and frequency of recreational drugs (if any); (6) Cravings relating to MDMA and any subsequent use of illicit ecstasy; and (7) Symptoms of AUD or any comorbid disorder(s). Those of skill in the art will appreciate that other equivalent or similar measures of therapeutic effect also may be used, and others are described herein and may be claimed. In some embodiments, a therapeutic effect may be shown by one or more of: (a) a reduction of alcohol use, (b) a reduction of alcohol cravings, (c) a promotion of alcohol abstinence, (d) a prevention of relapse into alcohol use, or (e) an improvement of at least one symptom of alcohol use disorder. In some embodiments, a therapeutic effect may be shown by one or more of: (a) an increase in quality of life, (b) an increase in psychosocial functioning, (c) a decrease in use or frequency of a prescription medication, (d) a decrease in use or frequency of a recreational drug, (e) a decrease in obsessive compulsive thoughts, (f) a decrease in suicidality, (g) an increase in feelings of empathy, or (h) an increase in self-compassion. In some embodiments, a therapeutic effect may be shown by an improvement of at least one symptom of a comorbid psychiatric disorder, such as antisocial personality disorder, borderline personality disorder, depression, anxiety, schizophrenia, attention deficit hyperactivity disorder, bipolar disorder, obsessive compulsive disorder, binge eating disorder, or PTSD. In some embodiments, a measure of therapeutic effect may be "durable,- e.g., where shown after a specific period of time, for example at least 1 month, at least 3 months, at least 6 months, at least 9 months, at least 1 year, or greater than I year, where such time is from the end of detoxification, if detoxification is completed, from baseline, or from the first psychotherapy session. In some embodiments, a measure of therapeutic effect will be shown for a period of time measured from when the subject has completed the MDMA-assisted psychotherapy regimen (i.e., measured from the last MDMA-assisted or non-drug assisted psychotherapy session).
[165] An "effective amount," a "therapeutically effective amount," a "therapeutically effective dose," or a "pharmacologically effective amount," refers to an amount of an active agent that is non-toxic and sufficient to provide the desired therapeutic effect with performance at a reasonable benefit/risk ratio attending any medical treatment. "[he effective amount will vary depending upon the subject and the disease condition being treated or health benefit sought, the weight and age of the subject, the severity of the disease condition or degree of health benefit sought, the manner of administration, and the like, all of which can readily be determined by one of skill. As it relates to the invention, "therapeutically effective amount,"
refers to the amount of a compound of the invention (i.e., MDMA) sufficient to effect treatment when administered to a subject in need of such treatment. Administration of a composition in a "therapeutically effective amount" or an "effective amount" to a subject means administration of an amount of the composition of the invention in a manner as disclosed herein, sufficient to achieve the desired effect. In some embodiments, a "therapeutically effective amount," or an "effective amount," thus would be the amount sufficient to cause a measurable modulation which, in some embodiments, refers to the exertion of a modifying or controlling influence on something which, as it relates to the invention, may be a neurotransmitter system (such as, but not limited to, the serotonergic, dopaminergic, and/or noradrenergic system), where that measurable modulation is known to result in or is shown to result in treatment of a disorder.
When an "effective amount" means an amount effective in treating the stated disorder itself, or in the symptoms thereof in a subject, "therapeutic effect" would be understood to mean the responses(s) in the subject after treatment judged to be desirable and beneficial.
[166] "Pharmacovigilance" refers to any efforts to monitor the physiological, psychological, or other effects associated with MDMA or any other active agent of the invention, especially in order to identify and evaluate adverse events or adverse reactions. For example, at every visit during a treatment period patients may be asked open-ended questions about how they are feeling and any side effects or adverse events documented. Therapists will have access to a check-list of common side effects to aid documentation.
[167] Commonly reported effects of MDMA reported in Phase 1 studies in healthy volunteers include elevation in blood pressure and heart rate, increased anxiety or dysphori a, and dilated pupils. Some reports indicated decreased rather than increased alertness Other common effects reported in controlled studies of MDMA include reduced appetite, dizziness, tight jaw or bruxism (tooth-grinding), difficulty concentrating, impaired gait or balance, dry mouth, and thirst. Patients in some studies also reported or exhibited changes in cognition, such as increases in speed of thought or thought blocking, facilitated imagination or facilitated recall, and unusual thoughts or ideas. Other less commonly reported effects include paraesthesias (unusual body sensations) such as tingling or feeling hot or cold. MDMA has been shown to produce anxiety in some healthy volunteers. These effects are transient and recede with the waning of drug effects. One study found that women were more likely than men to experience the most common effects of MDMA, though men were more likely than women to experience the specific effects of nausea and sweating. Effects in women undergoing a single session of MDMA-assisted psychotherapy for PTSD were mild and appear to be similar to those in healthy controls. Clinical judgment generally will be used to define any effects as adverse where appropriate and these will be documented using standard procedures, where such documentation is required or desired.
[168] "Adverse event" (AE) refers to any untoward medical occurrence in a patient or subject administered a medicinal product and which does not necessarily have a causal relationship with this treatment. An AE can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of an investigational medicinal product (IMP), whether or not considered related to the IMP. A
moderate rise in pulse and blood pressure is common when taking MDMA. For example, MDMA at doses of 100-125 mg has been associated with increases in heart rate of 26-30 bpm and blood pressure (diastolic 14.4-25 mmg) (Harris 2002, Mas 1999, Mithoefer et al. 2011).
These can be monitored following a regime such as that known in the art (see, e.g., Oehen et al. 2013, Chabrol & Oehen 2013). In some embodiments, blood pressure and pulse can be measured throughout a MDMA-assisted session, starting at baseline before the drug is given and hourly thereafter for six hours. In the event of a patient presenting with acutely elevated BP (e.g., systolic BP [SBP] >200 mm Hg or diastolic BP [DBP] >120 mm Hg) the patient is referred for emergency hospital care. The upper age range of patients may be limited, e.g., to 65 years, and patients may be screened for cardiac abnormalities.
[169] -Adverse reaction" (AR) refers to all untoward and unintended responses to an IMP
related to any dose administered. All ARs judged as having reasonable causal relationship to a medicinal product qualify as adverse reactions. The expression "reasonable causal relationship" means to convey in general that there is evidence or argument to suggest a causal relationship. As an example, there has been one serious adverse reaction associated with 125 mg of MDMA during assisted psychotherapy. This individual was observed to have an increased frequency of ventricular extrasystoles (PVCs) on the third of three MDMA-assisted sessions; this had not occurred on two previous sessions (after haying been given 125 mg and then 62.5 mg MDMA). The decision was taken not to administer the booster 62.5 mg MDMA
dose on this occasion and a single dose of a beta blocker was administered.
Monitoring as normal was undertaken with full recovery the following day. Extensive cardiac investigations revealed no evidence of persisting pathology. This patient was noted to have a family history of cardiovascular disease (biological father coronary artery bypass). There have been no other unexpected drug-related serious adverse events in any of the human MDMA
research studies to date (Doblin et al. 2014).
[170] "Unexpected adverse reaction" (UAR) refers to an AR, the nature or severity of which is not consistent with the applicable product information (e.g., investigator's brochure for an unapproved investigational product, summary of product characteristics (SmPC) for an authorized product, or package insert or other product labeling for an approved drug). When the outcome of the adverse reaction is inconsistent with applicable product information this adverse reaction should be considered -unexpected.- Side effects documented in the SmPC
which occur in a more severe form than anticipated are also considered to be unexpected.
[171] "Serious adverse event" (SAE) or "serious adverse reaction" (SAR) refer to any untoward medical occurrence or effect that at any dose: results in death; is life-threatening (i.e., an event in which the subject was at risk of death at the time of the event, but not an event which hypothetically might have caused death if it were more severe), requires hospitalization, or prolongation of existing inpatients' hospitalization; results in persistent or significant disability or incapacity; or is a congenital anomaly or birth defect. However, this list is non-exclusive, and medical judgment will be exercised in deciding whether an AE or AR is serious in other situations. Important AEs or ARs that are not immediately life-threatening or do not result in death or hospitalization but may jeopardize the subject or may require intervention to prevent one of the other outcomes listed in the definition above, also should be considered serious.
[172] "Suspected unexpected serious adverse reaction" (SUSAR) refers to any suspected adverse reaction related to an IMP that is both unexpected and serious (i.e., by reference to the definitions for UAR and SAR as disclosed herein)
[173] In discussing AEs and ARs as above, it will be appreciated that there are distinct differences between the uses of recreational ecstasy compared to clinical MDMA. The former is a drug preparation of varying quality and dose that commonly comprises one or more potential toxic adulterants. Although a number of serious adverse events, including fatalities, have been reported after recreational ecstasy use in unsupervised and uncontrolled settings, even these events are relatively rare given the prevalence of ecstasy use.
These include hyperthermia, including hyperthermia arising from "serotonin syndrome,"
psychiatric problems, hepatotoxicity, including acute liver failure arising from hyperthermia and liver disease and hyponatremia. Accordingly, in the compositions and methods of the invention, "MDMA," unless context expressly dictates otherwise, refers to MDMA that is pure or substantially pure, or a composition containing such pure or substantially pure MDMA (e.g., such MDMA plus an excipient, wherein "excipient," in some embodiments, refers to a substantially "inactive- substance that serves as the vehicle or medium for a drug or other substantially "active" substance, and an "inactive- substance refers to a substance that exerts substantially no effects on the body, while an "active" substance exerts observable¨through, among other metrics, objective measurements as disclosed herein¨effects on the body).
[174] "Set and setting" may play a role in the development of some ecstasy-related adverse events, such as vigorous physical exertion, lack of attention to somatic cues, and too little or too much hydration resulting in hyperthermia or hyponatremia. The term "set and setting" is commonly attributed to Timothy Leary, a psychologist well known for promoting psychedelics and psychedelic research in the 1960s. While consideration of set and setting is traced further back to use of plant-based psychedelics in Indigenous cultures, it continues to be considered to be crucial in modern use of psychedelic-assisted therapies in clinical settings and research trials (Hartogsohn, 2017). The concept of "set" refers to the internal aspects of the person undergoing a psychedelic experience such as their personality, mood, intentions and expectations (Metzer and Leary, 1967). "Setting" encompasses the physical, social and cultural environment in which the psychedelic experience takes place (Hartogsohn, 2017).
[175] These factors can be controlled during MDMA administration, whether by the supervising clinicians or a member of the therapy team, or by the design of the treatment protocol itself. Such factors are considered and controlled in the methods of the invention, as will become readily apparent. Both set and setting considerations are paramount to support an individual's therapeutic course. Careful preparation is crucial to allow a patient to feel ready to trust and let go in their drug-assisted session so that they can go towards and be with whatever internal experiences show up. In addition, supportive integration is fundamental to allow the individual to describe for themselves their internal experiences during the drug-assisted session and make meaning from these in ways that helps them take committed action towards their values. Integration goes beyond just the formal sessions that take place within the therapy setting, and a patient should be supported to consider how their ongoing processing can be facilitated via community networks and resources.
[176] Gaining a strong therapeutic alliance within a relatively short period of time can help the patient to feel supported and safe; using core therapeutic skills such as active listening, reflections using the patient's own words where appropriate and giving time to discuss any questions or anxieties will facilitate the patient-therapist relationship.
Wherever possible the therapist will attend the initial assessment to maximise the opportunity for rapport building, as well as beginning the early psychological formulation process. The assessment phase will help to determine whether a patient appears to be psychologically suited to the therapeutic intervention, notwithstanding other key eligibility criteria. Part of this consideration can take into account the person's support network within their community and their willingness and ability to utilize and/or build on this for support.
[177] The physical environment will be purposefully designed with safety and support in mind, while being minimally distracting to allow the patient to focus on their internal experience during drug-assisted sessions. Such design will be as readily appreciated in the art, and as used in psychedelic-assisted psychotherapy for many years. In line with both early and modern research with psychedelics, dating back at least decades, the therapy rooms are warm and comfortable spaces with furniture allowing the patient to recline with cushions and blankets if they choose. The room should be a quiet space with minimal distractions with the ability to play music both within the room and in the headphones simultaneously.
According to Kalean et al. (2018), this helps with: "(1) providing a sense of continuity in case the headphones are abruptly removed or muted, (2) enabling the therapists to empathise with the patient's current state, as well as observe how they were responding to a particular piece of music, and (3) allowing a deeper immersion in the music and depth of sound." Music is a well-established feature of psychedelic assisted therapies and is described by Kalean et al.
(2018) as "the hidden therapist" due to its potential to impact upon the individual's internal experience in meaningful ways while under the influence of a mind-altering compound.
[178] "Causality" and "causal relationship" for an AE also may be assessed, i.e., an AE may be assessed as to whether or not there is a reasonable possibility of a causal relationship to the composition and/or its administration or method of use (e.g., a protocol of MDMA-assisted psychotherapy). In any large group of patients having AUD, a large number of AEs are expected due to the nature of the patients' condition. As the intervention is composed of psychotherapy sessions as well as MDMA-assisted psychotherapy sessions, those performing the methods herein will in some embodiments document causality according to intervention where possible. The assignment of the causality may be made by the physician, investigator, clinical psychologist, or other member of the medical or treatment team responsible for the care of the patient using the definitions in TABLE 3 below. On occasion, doubt about the causality may exist. In such circumstances, all members of the medical or treatment team can speak together or with other knowledgeable clinicians. In the case of continued discrepant views, additional outside clinicians can be asked to advise.
TABLE 3: Definitions Regarding the Assignment of Causality to an AE
Relationship Description Unrelated There is no evidence of any causal relationship Unlikely There is little evidence to suggest there is a causal relationship (e.g., the event did not occur within a reasonable time after administration of the medication).
There is another reasonable explanation for the event (e.g., the patient's clinical condition, other concomitant treatment).
Possible There is some evidence to suggest a causal relationship (e.g., because the event occurs within a reasonable time after administration of the medication).
However, the influence of other factors may have contributed to the event (e.g., the patient's clinical condition, other concomitant treatments).
Probable There is evidence to suggest a causal relationship and the influence of other factors is unlikely.
Definitely There is clear evidence to suggest a causal relationship and other possible contributing factors can be ruled out.
Not There is insufficient or incomplete evidence to make a clinical judgment of the assessable causal relationship.
[179] "Psychedelic-assisted psychotherapy" refers to a range of related approaches that involve at least one session where the patient ingests a "psychedelic" and is monitored, supported, and/or otherwise engaged by one or more trained mental health professionals while under the effects of the drug (see, e.g., Schenberg 2018). Protocols have been developed for the standardization of procedures which emphasize a high degree of care (see, e.g., Johnson 2008), such as the therapeutic approach used by MAPS to treat patients with PTSD using MDMA (e.g., described in Mithoefer 2017). It will be appreciated that when the term "psychedelic-assisted psychotherapy" is used, the drug compound in the psychotherapy need not be a "psychedelic" as the class is sometimes understood (e.g., the class of "classic"
psychedelics), but also may include drugs having "entactogenic" or "empathogenic" effects like MDMA, or drugs having "dissociative" effects such as ketamine and esketamine.
[180] A variety of methods of psychedelic-assisted psychotherapy will be known to those in the field and are disclosed, for example, in Grof 2008; Passie 2012; Johnson et al. 2008; Sessa & Fischer 2015; Schmid et al. 2020; Greer & Tolbert 1998; Mithoefer 2017;
Mithoefer 2013;
U.S. Pat. App. No. 2020/0360311A1, discussing drug-assisted psychotherapy practices.
[181] "Additional therapeutic effects" that may be increased in some embodiments of the invention include, but are not limited to, antioxidant, anti-inflammatory, analgesic, antineuropathic, antinociceptive, antimigraine, anxiolytic, antidepressant, antipsychotic, anti-PTSD, immunostimul ant, anti-cancer, anti emetic, orexigenic, antiulcer, antihistamine, antihypertensive, anticonvul sant, anti epileptic, bronchodilator, neuroprotective, entactogenic, empathogenic, entheogenic, euphoric, psychedelic, sedative, and stimulant effects.
[182] "Synergistic effects" will be understood as including increases in potency, bioactivity, bioaccessibility, bioavailability, or therapeutic effect (including one or more additional therapeutic effects), that are greater than the additive contributions of the components acting alone, and/or are greater than the contribution of the isolated compounds on their own. In some embodiments, "synergistic" refers to a pharmaceutical composition or combination therapy that is more effective than the additive effects of any two or more single agents. A synergistic effect, for example, permits the effective treatment of a disease using lower amounts (doses) of individual therapy. This includes lower doses of the first pharmaceutical agent or the second pharmaceutical agent ("apparent one-way synergy"), or lower doses of both pharmaceutical agents (-two-way synergy"), than would normally be required when either drug is used alone.
In effect, the lower doses result in lower toxicity without reduced efficacy.
A synergistic effect may additionally result in improved efficacy, including an improved avoidance or reduction of disease as compared to any single therapy.
[183] "Synergistic effects" will also be understood to include increases in potency, bioactivity, bioaccessibility, bioavailability, or therapeutic effect (including one or more additional therapeutic effects), that are greater than the additive contributions of the components acting alone, and/or are greater than the contribution of the isolated compounds on their own. Numerous methods known to those of skill in the art exist to determine whether there is synergy as to a particular effect, i.e., whether, when two or more components are mixed together, the effect is greater than the sum of the effects of the individual components applied alone, thereby producing "1+1 = 2." Suitable methods include isobologram (or contour) analysis (Huang et al. 2019), or the equation of Loewe additivity (Loewe &
Muischnek, 1926, Arch. Exp. Pathol Pharmacol. 114: 313-326). A synergistic effect also may be calculated using methods such as the Sigmoid-Emax equation (Holford & Scheiner, 1981, Clin.
Pharmacokinet.

6: 429-453) and the median-effect equation (Chou & Talalay, 1984, Adv. Enzyme Regul.
22:27-55). The corresponding graphs associated with the equations referred to above are the concentration-effect curve and combination index curve, respectively. Each equation referred to above may be applied to experimental data to generate a corresponding graph to aid in assessing the effects of the drug combination.
[184] The goal of increasing an existing therapeutic effect, providing an additional therapeutic effect, increasing a desired property such as stability or shelf-life, decreasing an unwanted effect or property, altering a property in a desirable way (e.g., pharmacoki nett cs or pharmacodynamics), modulating a desired system or pathway (e.g., a neurotransmitter system), or otherwise inducing synergy, also can be achieved by the inclusion of an additional active compound. Such additional active compounds may include any one or more of amino acids, antioxidants, anti-inflammatory agents, analgesics, antineuropathic and antinociceptive agents, antimigraine agents, anxiolytics, antidepressants, antipsychotics, anti-PTSD
agents, immunostimulants, anti-cancer agents, antiemetics, orexigenics, antiulcer agents, antihistamines, antihypertensives, anticonvulsants, antiepileptics, bronchodilators, neuroprotectants, other entactogens and empathogens, entheogens, psychedelics, monoamine oxidase inhibitors, sedatives, stimulants, and vitamins. These ingredients may be in ion, freebase, or salt form, include polymorphs, and may be isomers.
[185] In some embodiments, an additional active agent may be any one or more of an opioid antagonist (e.g., nalmefene, naltrexone), a CBI antagonist (e.g., rimonabant), a CRH1 receptor antagonist (e.g., verucerfont, pexacerfont), a NKIR antagonist (e.g., tradipitant), an OTR
agonist (e.g., oxytocin), a GABA agent (e.g., topiramate, baclofen, a benzodiazepine, such as alprazolam, diazepam or lorazepam), a voltage-gated sodium channel inhibitor (e.g., oxacarbazepine, valproic acid, zonisamide), a voltage-dependent calcium channel agonist (e.g., gabapentin, pregabalin), an a7 nicotinic acetylcholine receptor agonist (e.g., varenicline), a 5-HT3 antagonist (e.g., ondansetron), a 5-HT1A, receptor partial agonist (e.g., aripiprazole), a 5-HT2A receptor antagonist (e.g., quetiapine, olanzapine, mirtazapine), a 5-HT
reuptake inhibitor (e.g., trazodone), a SERT inhibitor (e.g., duloxetine), a al adrenoreceptor antagonist (e.g., doxazosin, prazosin), a glucocorticoid receptor antagonist (e.g., mifepristone), an al adrenoreceptor agonist (e.g., guanfacine), an AChE inhibitor (e.g., citicoline), a dopamine D2 receptor antagonist (e.g., tiapride), an a2 adrenoreceptor agonist (e.g., clonidine), an NMDA
receptor antagonist (e.g., acamprosate) and an aldehyde dehydrogenase inhibitor (e.g., disulfiram), or pharmaceutically acceptable salts thereof.
[186] The compounds described herein may contain one or more asymmetric centers and give rise to enantiomers, diastereomers, and other stereoisomeric forms. Each chiral center may be defined, in terms of absolute stereochemistry, as (R)- or (S)-. The invention is meant to include all such possible isomers, as well as mixtures thereof, including racemic and optically pure forms. Thus, for example, chiral compounds will be understood to refer to racemic mixtures, as well as any of the individual enantiomers, and all enantiomerically enriched mixtures thereof, in any proportions. Optically active (R)- and (S)-, (-)- and (+)-, or (D)- and (L)-isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. Various methods are known in the art for preparing optically active forms and determining activity. Such methods include standard tests described herein and other similar tests which are well known in the art.
[187] The compounds of the invention may be provided in a composition that is enantiomerically enriched, such as a mixture of enantiomers in which one enantiomer is present in excess, including an enantiomeric excess in any amount greater than 0%, and in some embodiments in an enantiomeric excess of at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, at least about 99.5%, at least about 99.9%, up to (and including) 100%. In some embodiments are enantiomerically enriched mixtures in an enantiomeric excess of less than about 50%, such as less than about 45%, less than about 40%, less than about 35%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, less than about 5%, including less than about 1%, less than about 0.5%, and up to 0%.
[188] In some embodiments as described above, e.g., a mixture with an enantiomeric excess of at least about 50% or less than about 50%, the compound will be MDMA. In some embodiments are compounds, e.g., MDMA, which are present in a composition or used in a method, and which are a pure or substantially pure individual enantiomer, such as a mixture having an enantiomeric excess of greater than about 75%, about 76%, about 77%, about 78%, about 79%, about 80%, about 81%, about 82%, about 83%, about 84%, about 85%, about 86%, about 87%, about 88%, about 89%, or preferably above about 90%, such as about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, about 99.5%, about 99.9%, and up to about 100% and including 100%.
[189] When any compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, tautomeric forms are included.
[190] The individual compounds of the compositions of the invention will be understood to also encompass pharmaceutically acceptable salts of such compounds. The term "pharmaceutically acceptable salt" refers to salts prepared from pharmaceutically acceptable non-toxic acids or bases, and which may be synthesized by conventional chemical methods.
Generally, such salts are prepared by reacting the free acid or base forms of these agents with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media (e.g., ether, ethyl acetate, ethanol, isopropanol, or acetonitrile) are preferred. For therapeutic use, salts of the compounds are those wherein the counter-ion is pharmaceutically acceptable. Exemplary salts include, but are not limited to, 2-hy droxy ethane s ul fonate, 2-naphth al enesulfonate, 2-nap sylate, 3 -hy droxy -2-naphthoate, 3-phenylpropionate, 4-acetamidobenzoate, acefyllinate, acetate, aceturate, adipate, alginate, aminosalicylate, ammonium, amsonate, ascorbate, aspartate, benzenesulfonate, benzoate, besylate, bicarbonate, bisulfate, bitartrate, borate, butyrate, calcium edetate, calcium, camphocarbonate, camphorate, camphorsulfonate, camsylate, carbonate, cholate, citrate, cl avul ari ate, cy cl op entanepropi onate, cypionate, d-aspartate, d-cam syl ate, d-lactate, decanoate, dichloroacetate, digluconate, dodecylsulfate, edentate, edetate, edisylate, estolate, esylate, ethanesulfonate, ethyl sulfate, fumarate, furate, fusidate, galactarate (mucate), galacturonate, gallate, gentisate, gluceptate, glucoheptanoate, gluconate, glucuronate, glutamate, glutarate, gl y ceroph o sph ate, glycol ate, glycollylarsani late, hemi sulfate, heptanoate (enanthate), heptanoate, hexafluorophosphate, hexanoate, hexylresorcinate, hippurate, hybenzate, hydrabamine, hydrobromide, hydrobromide/bromide, hydrochloride/chloride, hy drofluori de/fluori de, hydroi odi de, hydroxide, hydroxybenzoate, hydroxynaphthoate, iodide, isethionate, isothionate, 1-aspartate, 1-camsylate, 1-lactate, lactate, lactobionate, laurate, laurylsulphonate, lithium, magnesium, malate, maleate, malonate, mandelate, meso-tartrate, mesylate, methanesulfonate, methylbromide, methylnitrate, methylsulfate, mucate, myristate, N-methylglucamine ammonium salt, napadisilate, naphthylate, napsylate, nicotinate, nitrate, octanoate, ol eate, orotate, ox al ate, p-toluenesul fon ate, palm i tate, p am oate, p antoth en ate, pectinate, p ersul fate, phenyl propi on ate, phosphate, phosphate] di phosphate, pi crate, pi val ate, polygalacturonate, potassium, propionate, pyrophosphate, saccharate, salicylate, salicylsulfate, sodium, stearate, subacetate, succinate, sulfate, sulfosaliculate, sulfosalicylate, suramate, tannate, tartrate, teocl ate, terephthal ate, thiocyanate, thi o s al i cyl ate, to syl ate, trib rophen ate, triethiodide, undecanoate, undecylenate, valerate, valproate, xinafoate, zinc and the like, as would be immediately evident to one of skill (Berge et al. 1977). Preferred pharmaceutically acceptable salts, where the compound is MDMA, include those employing a hydrochloride anion (e.g., an HC1 salt).
[191] Different embodiments of the invention include the following examples:
Pharmaceutically acceptable complex derivatives of each drug in each group, including solvates, salts, esters, enantiomers, isomers (stereoisomers and/or constitutional, including, but not limited to, ones based on substituting deuterium or a halogen, such as bromine, chlorine, fluorine, and iodine; for any hydrogen), derivatives or prodrugs of the compounds of the invention. Prodrugs of the active agents also will be appreciated to be within the scope of the invention. The term "prodrug" refers to a precursor of a biologically active pharmaceutical agent. Prodrugs undergo a chemical or a metabolic conversion to become a biologically active pharmaceutical agent. A prodrug can be converted ex vivo to the biologically active pharmaceutical agent by chemical transformative processes. In vivo, a prodrug is converted to the biologically active pharmaceutical agent by the action of a metabolic process, an enzymatic process or a degradative process that removes the prodrug moiety, such as a glycoside or acetyl group, to form the biologically active pharmaceutical agent. Other examples include addition of hydroxy groups (Tsujikawa et al 2011. Xenobiotica, 41(7), 578-584; Yamamoto et al. 1984.
Xenobiotica, 14(11), 867-875), acyloxyalkoxycarbonyl derivatives, amino acids, or peptides (Vig et al. 2013. Advanced Drug Delivery Reviews, 65(10), 1370-1385), which are generally added to the amine, and can be removed within the body by chemical reactions or enzymes, but other prodrugs and precursors, at the amine and other sites, should be understood to be within the scope of the invention (Simplicio, Clancy, & Gilmer. 2008.
Molecules, 13(3), 519-547; Shah, Chauhan, Chauhan, & Mishra (Eds.). 2020. Recent Advancement in Prodrugs. CRC
Press). Types of prodrugs contemplated to be within the scope and spirit of the invention therefore include compounds that are transformed in various organs or locations in the body (e.g., liver, kidney, GI., lung, tissue) to release the active agent. For example, liver prodrugs will include active agents conjugated with a polymer or chemical moiety that is not released until acted upon by liver cytochrome enzymes; CYP metabolism includes dealkylati on, dehydrogenation, reduction, hydrolysis, oxidation, and the breakdown of aromatic rings.
Kidney prodrugs will include active agents conjugated to L-gamma-glutamyl or N-acetyl-L-gamma glutamic moieties so that they are metabolized by gamma-glutamyl transpeptidase before they are bioactive; alternatively, they may be conjugated to alkylglucoside moieties to create glycosylation-based prodrugs. Digestive or G.I. prodrugs will include those where an active agent is, e.g., formulated into microspheres or nanospheres that do not degrade until the spheres are subjected to an acidic pH; formulated with an amide that will resist biochemical degradation until colonic pH is achieved; or conjugated with a linear polysaccharide such as pectin that will delay activation until the pharmaceutical composition reaches the bacteria in the colon. Besides such exemplary prodrug forms, many others will be known to one of skill.
[192] Among derivatives of a compound are included its "physiologically functional derivatives," which refers to physiologically tolerated chemical derivatives of the compound having the same physiological function thereof, for example by being convertible in the body thereto, and which on administration to a mammal such as a human is able to form (directly or indirectly) the compound or an active metabolite thereof (acting therefore, like a prodrug), or by otherwise having the same physiological function, despite one or more structural differences. Meaning, a therapeutically effective dose of a prodrug would necessarily be a therapeutically effective dose of the corresponding biologically-active compound. Examples of physiologically functional derivatives may include esters (including diacid hemiesters), amides, carbamates, ureas, and heterocycles.
[193] In some embodiments, the compounds of the invention are synthetic. -Synthetic"
means a compound which is manufactured artificially in a laboratory, by means of chemical synthesis (e.g., by a series of chemical processes or reactions using chemical substrates, reagents, and optionally one or more catalysts) or biosynthesis (e.g., from a bioengineered organism, and thus including those compounds also referred to as "biosynthetic" or as involving "synthetic biology" or "synbio"). For example, bacteria, yeast, and other host cells can be bioengineered to produce synthetic compounds by inserting genes that produce appropriate enzymes and/or by altering the natural metabolic pathway to achieve the production of the desired compound(s), e.g., from sugars, the main carbon source available to yeast, or from other precursor molecules. Cell-free protein synthesis (CFPS) and in vitro protein synthesis are also contemplated. These compounds can then be obtained and purified.
[194] Generally, the individual compounds of the invention shall be administered as part of a pharmaceutical composition or formulation, but will be prepared for inclusion in such composition or formulations as isolated or purified compounds. In certain preferred embodiments, a compound of the invention is in a substantially pure or isolated preparation.

The terms "isolated," "purified," or "substantially pure" as used herein, refer to material that is substantially or essentially free from components that normally accompany the material when the material is synthesized, manufactured, or otherwise produced, according to a particular degree of purity, and when measured by a method described herein or of general knowledge in the art. Any compound of the disclosure can be purified. A compound (or a composition comprising a compound) as described herein will generally be at least about 90% pure, including at least about 95% pure, at least about 96% pure, at least about 97%
pure, at least about 98% pure, at least about 99% pure, at least about 99.1% pure, at least about 99.2% pure, at least about 99.3% pure, at least about 99.4% pure, at least about 99.5%
pure, at least about 99.6% pure, at least about 99.7% pure, at least about 99.8% pure, at least about 99.9% pure, or up to 100% pure. A "substantially pure" compound (or a composition comprising a compound) may be defined as a compound or composition having a chromatographic purity (of the desired compound) of greater than about 90%, more preferably greater than about 95%, more preferably greater than about 96%, more preferably greater than about 97%, more preferably greater than about 98%, more preferably greater than about 99%, more preferably greater than about 99.25%, more preferably greater than about 99.50%, more preferably greater than about 99.75%, and most preferably greater than about 99.90%, greater than about 99.95%, or greater than about 99.99%, as determined by area normalization of HPLC profile or other similar detection method.
[195] A substantially pure compound of the invention is, preferably, substantially free of any other active agents which are not intended to be administered to a subject. In this context "substantially free" can be taken to mean that no impurities, adulturants, or active agent(s) other than the active agent intended to be administered to a subject are detectable by HPLC or other similar detection method, or are below a desired threshold of detection such as defined above. Other active agents that may be found in a compound that is not substantially pure include those known in the art to be present in recreational or non-clinical MDMA, such as by-products and side-products of non-GMP synthesis, or compounds sold as or with -MDMA"
while not in fact MDMA (e.g., other phenethylamines, cathinones, etc.) (see, e.g., Saleemi 2017). In some embodiments, the composition of the invention therefore will comprise less than about 5% impurities or adulterants, including additional (undesired) active agents such as described above, and in some embodiments will comprise less than 4%, less than 3%, less than 2%, less than 1%, less than 0.5%, less than 0.4%, less than 0.3%, less than 0.2%, less than 0.1%, less than 0.05%, less than 0.025%, or less than 0.01% impurities or adulterants, including in some embodiments no measurable impurities or adulterants.
B. Compositions and Formulations
[196] "Pharmaceutical compositions" are compositions comprising disclosed compound(s) together in an amount (for example, in a unit dosage form) with a pharmaceutically acceptable carrier, diluent, or excipient It should be understood that some embodiments do not have a single carrier, diluent, or excipient alone, but include multiple carriers, diluents, and/or excipients. Compositions can be prepared by standard pharmaceutical formulation techniques such as disclosed in Remington: The Science and Practice of Pharmacy (2005) 21th ed., Mack Publishing Co., Easton, Pa.; The Merck Index (1996) 12th ed., Merck Publishing Group, Whitehouse, N.J., Pharm. Principles of Solid Dosage Forms (1993), Technomic Publishing Co., Inc., Lancaster, Pa.; and Ansel and Stoklosa, Pharm. Calculations (2001) 11th ed., Lippincott Williams & Wilkins, Baltimore, Md.; and Poznansky et al. Drug Delivery Systems (1980), R.L. Juliano, ed., Oxford, N.Y., pp. 253-315).
[197] "Pharmaceutically acceptable" as used in connection with one or more ingredients means that the ingredients are generally safe and, within the scope of sound medical judgment, suitable for use in contact with the cells of humans and other animals without undue toxicity, irritation, allergic response, or complication, and commensurate with a reasonable risk! benefit ratio.
[198] These compositions can be prepared as formulations suitable for administration by a variety of routes, non-limiting examples of which include enteral means, wherein "enteral means" or "enteral administration means" includes, but is not limited to oral solid and oral liquid dosage forms, sublingual, and buccal administration means; as well as parenteral means, wherein "parenteral means" or -parenteral administration means" includes but is not limited to, intravenous, intra-arterial, intraperitoneal, intraosseous, intramuscular, intrathecal, intracerebroventricular, rectal, vaginal, ocular, nasal, cutaneous, topical, otic, ocular, transdermal, and subcutaneous administration means. As appreciated by one of skill, the compounds employed in the methods of the invention are effectively administered as oral solid and oral liquid dosage forms; sublingually or buccally; as injections, including intravenous, intra-arterial, intraperitoneal, intraosseous, intramuscular, intrathecal, and intracerebroventricular; rectally, vaginally, ocularly, nasally, cutaneously, topically, oticly, transdermally, and subcutaneously. Additionally, the compounds employed in the methods of the invention are effectively administered via other means, and prepared as any acceptable compositions known to those of skill. Such compositions may be prepared in any manner known in the pharmaceutical arts that comprise at least one active agent (Sheth et al., 1980).
[199] In making the compositions employed in the present invention the active ingredients are often mixed with an excipient, diluted by an excipient, or enclosed within such a carrier which can be in the form of a capsule, sachet, paper, or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier, or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments, soft or hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
Different embodiments include immediate, delayed, extended, and controlled release forms. Many other variations are possible and known to those skilled in the art.
[200] In some embodiments of the invention are multiple routes of administration, which may differ between patients according to patient preferences, comorbidities, side effect profiles, pharmacokinetic and pharmacodynamic considerations, and other factors. In some embodiments are the presence of other substances with the active agents, known to those skilled in the art, such as fillers, carriers, gels, skin patches, lozenges, or other modifications in the preparation to facilitate absorption through various routes (e.g., gastrointestinal, transdermal, etc.), to extend the effect of the drugs, and/or attain higher or more stable serum levels or enhance the therapeutic effect of the active agents in the combination.
[201] In preparing a formulation, it may be necessary to mill the active agent to provide the appropriate particle size prior to combining with the other ingredients. If an active agent is substantially insoluble, it ordinarily is milled to a particle size of less than about 200 mesh.
[202] Excipients, as defined herein, may include, but are not limited to, fillers, diluents, lubricants, surfactants, glidants, binders, dispersing agents, suspending agents, disintegrants, viscosity-increasing agents, film-forming agents, granulation aid, flavoring agents, sweetener, coating agents, solubilizing agents, and combinations thereof.
[203] The pharmaceutical compositions described herein can be formulated into any suitable dosage form, including aqueous oral dispersions, aqueous oral suspensions, solid dosage forms including oral solid dosage forms, aerosols, controlled release formulations, fast melt formulations, effervescent formulations, self-emulsifying dispersions, solid solutions, liposomal dispersions, lyophilized formulations, tablets, capsules, pills, powders, delayed-release formulations, immediate-release formulations, modified release formulations, extended-release formulations, pulsatile release formulations, multi particulate formulations, and mixed immediate release and controlled release formulations. Generally speaking, one will desire to administer an amount of the active agent of the invention that is effective to achieve a plasma level commensurate with the concentrations found to be effective in vivo for a period of time effective to elicit the desired therapeutic effect(s).
[204] In some embodiments, the compositions are formulated in a unit dosage form. The term "unit dosage form" refers to a physically discrete unit suited as unitary dosages for the subject to be treated, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect(s), in association with a suitable pharmaceutical carrier, diluent, or excipient. Unit dosage forms are often used for ease of administration and uniformity of dosage. Unit dosage forms can contain a single or individual dose or unit, a sub-dose, or an appropriate fraction thereof (e.g., one half a -full" dose), of the pharmaceutical composition administered. Unit dosage forms include capsules, troches, cachets, lozenges, tablets, ampules and vials, which may include a composition in a freeze-dried or lyophilized state; a sterile liquid carrier, for example, can be added prior to administration or delivery in vivo. Unit dosage forms also include ampules and vials with liquid compositions disposed therein. Unit dosage forms further include compounds for transdermal administration, such as "patches" that contact the epidermis of a subject for an extended or brief period of time.
[205] In some embodiments, the compositions of the invention are formulated in a pharmaceutically acceptable oral dosage form. Oral solid dosage forms may include but are not limited to, lozenges, troches, tablets, capsules, caplets, powders, pellets, multiparticulates, beads, spheres, and/or any combinations thereof. Oral solid dosage forms may be formulated as immediate release, controlled release, sustained release, extended release, or modified release formulations.
[206] In some embodiments, the oral solid dosage forms of the invention may be in the form of a tablet (including a suspension tablet, a fast-melt tablet, a bite-disintegration tablet, a rapid-disintegration tablet, an effervescent tablet, or a caplet), a pill, a powder (including a sterile packaged powder, a dispensable powder, or an effervescent powder), a capsule (including both soft or hard capsules, e.g., capsules made from animal-derived gelatin or plant-derived HPMC, or "sprinkle capsules"), solid dispersion, solid solution, bioerodible dosage form, controlled release formulations, pulsatile release dosage forms, multiparticulate dosage forms, pellets, granules, or an aerosol. In other embodiments, the pharmaceutical formulation is in the form of a powder. In still other embodiments, the pharmaceutical formulation is in the form of a tablet, including a fast-melt tablet. Additionally, pharmaceutical formulations of the invention may be administered as a single capsule or in multiple capsule dosage form. In some embodiments, the pharmaceutical formulation is administered in two, three, four, or more capsules or tablets.
[207] Oral solid dosage forms may contain pharmaceutically acceptable excipients such as fillers, diluents, lubricants, surfactants, glidants, binders, dispersing agents, suspending agents, disintegrants, viscosity-increasing agents, film-forming agents, granulation aid, flavoring agents, sweetener, coating agents, solubilizing agents, and combinations thereof. Oral solid dosage forms also can comprise one or more pharmaceutically acceptable additives such as a compatible carrier, complexing agent, ionic dispersion modulator, disintegrating agent, surfactant, lubricant, colorant, moistening agent, plasticizer, stabilizer, wetting agent, anti-foaming agent, alone or in combination, as well as supplementary active agent(s).
Supplementary active agents include preservatives, antioxidants, antimicrobial agents including biocides and biostats such as antibacterial, antiviral and antifungal agents.
[208] In some embodiments, the pharmaceutical composition may be an "immediate release formulation," wherein a therapeutically effective amount of the pharmaceutical composition is administered to the subject in a way that facilitates rapid release. Immediate-release formulations may be prepared by combining a superdisintegrant such as croscarmellose sodium and different grades of microcrystalline cellulose in different ratios In some embodiments, to aid disintegration, sodium starch glycolate may be added.
[209] Tablets of the invention can be prepared by methods well known in the art. Various methods for the preparation of the immediate release, modified release, controlled release, and extended-release dosage forms (e.g., as matrix tablets having one or more modified, controlled, or extended-release layers) and the vehicles therein are well known in the art. For example, a tablet may be made by compression or molding. Compressed tablets may be prepared by compressing, in a suitable machine, an active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface-active or dispersing agent. Molded tablets may be produced by molding, in a suitable apparatus, a mixture of powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide a slow or controlled release of the active ingredient therein. Generally recognized compendia of methods include Remington 2020 and Sheth et at. 1980.
[210] In some embodiments, solid dosage forms are prepared by mixing the active agents of the invention with one or more pharmaceutical excipients to form a "bulk blend" composition_ The bulk blend composition is homogeneous, i.e., the active agents are dispersed evenly throughout so that the bulk blend may be readily subdivided into equally effective unit dosage forms, such as tablets, pills, and capsules. The individual unit dosages may also comprise film coatings, which disintegrate upon oral ingestion or upon contact with diluents. These formulations can be manufactured by conventional pharmaceutical techniques.
Conventional pharmaceutical techniques for preparation of solid dosage folins include, but are not limited to, the following methods, which may be used alone or in combination: (1) dry mixing, (2) direct compression, (3) milling, (4) dry or non-aqueous granulation, (5) wet granulation, or (6) fusion (see, e.g., Lachman et al. 1986). Other methods include spray drying, pan coating, melt granulation, granulation, fluidized bed spray drying or coating (e.g., Wurster coating), tangential coating, top spraying, tableting, and extruding.
[211] Depending on the desired release profile, oral solid dosage forms may be prepared as immediate release formulations, or as modified release formulations, such as controlled release, extended release, sustained release, or delayed release. In some embodiments, the release profile is determined by obtaining objective measurements from a patient, the objective measurements including but are not limited to, weight, body temperature, heart rate, respiratory rate, blood oxygenation, BP and its variables, including, but not limited to:
SBP, SBP, MAP, and PP; CNIBP; ECG measurements, including RR interval or its variability, QT
interval or its variability, I-ERV (including measured by devices other than an ECG);
hemodynamic response, and levels of glucose, cortisol, serotonin, dopamine, cholesterol; EEG
measures; BDNF;
genetic markers including relating to CYP enzymes or drug metabolism; genetic variation in mGluR5 or FKBP5; and numerous more, as will be readily appreciated by those of skill.
[212] In some embodiments of modified release formulations, the half-life compared to the half-life of an immediate release formulation is greater by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 50%, at least 75%, at least 100%, or values in between. In some embodiments of modified release formulations, the formulations are designed to result in a comparable "area under the curve," or AUC0_24, and a similar safety and efficacy profile, but having a delayed time to maximum concentration (tmax) of at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 50%, at least 75%, at least 100%, or values in between, as would be appreciated by one of skill. In some preferred embodiments, a formulation is designed to be a product with a specific time course based on an optimum -therapeutic window," such as less than about 30 minutes, about 30 minutes, about 45 minutes, about 60 minutes, about 90 minutes, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, and greater than 8 hours, including lengths of time in between.
[213] In some embodiments, oral solid dosage forms are formulated as a delayed release dosage form by utilizing an enteric coating to affect release in the small intestine of the gastrointestinal tract. An enteric-coated oral dosage form may be a compressed or molded or extruded tablet/mold (coated or uncoated) containing granules, powder, pellets, beads or particles of the active ingredient and/or other composition components, which are themselves coated or uncoated. The enteric-coated oral dosage form may also be a capsule (coated or uncoated) containing pellets, beads or granules of the solid carrier or the composition, which are themselves coated or uncoated. In some embodiments, enteric coatings may be used to prepare other controlled release dosage forms, including but not limited to extended release and pulsatile release dosage forms. Pulsatile release dosage forms may be formulated using techniques known in the art, such as those described in U.S. Pat. Nos.
5,011,692, 5,017,381, 5,229,135, and 5,840,329. Other suitable dosage forms are described in U.S.
Pat. Nos.
4,871,549, 5,260,068, 5,260,069, 5,508,040, 5,567,441 and 5,837,284.
[214] In some embodiments, the controlled release dosage form is a pulsatile release solid oral dosage form comprising at least two groups of particles, each containing active agents of the invention. "Particles" refers to homogenous groupings of substantially the same active agent. The first group of particles, upon ingestion by a subject, provides a substantially immediate dose of the active agents of the invention, and may either be uncoated, or comprise a coating and/or sealant. Using such means, a single unit dosage form can provide both a first and a second dosage amount in the single form (i.e., a first dosage amount in an immediate release form, and a second dosage amount in a delayed release form). In some embodiments, gastroretentive sustained release tablets are formulated by using a combination of hydrophilic polymer (e.g., hydroxypropyl methylcellulose), together with at least one swelling agent (e.g., crospovidone, sodium starch glycolate, and croscarmellose sodium), and an effervescent substance (e.g., sodium bicarbonate). Using known methods, gastroretentive tablets can be formulated to prolong the gastric emptying time and extend the mean residence time (MRT) in the stomach for optimal drug release and absorption (see, e.g., Arza 2009).
Coatings for providing a controlled, delayed, or extended release may be applied to the compositions of the invention or to a core containing the compositions, and may comprise a pharmaceutically acceptable ingredient in an amount sufficient to provide a delayed release from, for example, about 1 hour to about 7 hours following ingestion before release of the active agents. Suitable coatings include one or more differentially degradable coatings including pH-sensitive coatings (enteric coatings), or non-enteric coatings having variable thickness to provide differential release of the active agents. Many other types of modified release systems will be known to those of skill. Non-limiting examples of additional delivery systems include both polymer- and nonpolymer-based systems, silastic systems, peptide-based systems, wax coatings, bioerodible dosage forms, and compressed tablets using conventional binders (see, e.g., Liberman et al.
1990; Singh et al. 2002; U.S. Pat. Nos. 4,327,725; 4,624,848; 4,968,509;
5,461,140; 5,456,923;
5,516,527; 5,622,721; 5,686,105; 5,700,410; 5,977,175; 6,465,014; and 6,932,983).
[215] In some embodiments, the compositions of the invention are formulated as a pharmaceutically acceptable oral liquid form. Oral liquid dosage forms include tinctures, drops, emulsions, syrups, elixirs, suspensions, and solutions, and the like. These oral liquid dosage forms may be formulated with any pharmaceutically acceptable excipient known to those of skill for the preparation of liquid dosage forms, and with solvents, diluents, carriers, excipients, and the like chosen as appropriate to the solubility and other properties of the active agents and other ingredients. Solvents may be, e.g., water, glycerin, simple syrup, alcohol, medium chain triglycerides (MCT), and combinations thereof.
[216] Oral liquid dosage forms are especially advantageous in treating geriatric and pediatric populations, as the liquid formulation is much easier to swallow. The liquid formulation is also absorbed more rapidly than a solid dosage form, as it need not be dissolved by the digestive system before it is available for absorption.
[217] Oral liquid dosage forms may be monophasic or biphasic, the former being a substantially homogenous solution dissolved in water or non-aqueous solvent, while the latter refers to oral liquid dosage forms in which the active ingredients do not fully dissolve in common solvents. Thus, over time, the solid particles (i.e., the active agents) within the oral liquid dosage form may form a precipitate at the bottom of the container¨requiring vigorous shaking to redisperse the active ingredients. Examples of monophasic liquid forms include syrups, linctuses, spirits/essences, elixirs, and fluid extracts. Examples of biphasic liquid forms include oral suspensions, oral emulsions, and mixtures.
[218] Liquid dosage forms for oral administration may be prepared as liquid suspensions or solutions using a sterile liquid, such as but not limited to, an oil, water, an alcohol, combinations of pharmaceutically suitable surfactants, suspending agents, and emulsifying agents Liquid formulations also may be prepared as single dose or multi-dose beverages.
Suspensions may include oils. Such oils include, but are not limited to peanut oil, sesame oil, cottonseed oil, corn oil, and olive oil. Suitable oils also include carrier oils such as MCT and long chain triglyceride (LCT) oils. Suspension preparation may also contain esters of fatty acids such as ethyl oleate, isopropyl myristate, fatty acid glycerides, and acetylated fatty acid glycerides. Suspension formulations may include alcohols, such as ethanol, isopropyl alcohol, hexadecyl alcohol;
glycerol, and propylene glycol. Ethers, such as polyethylene glycol; petroleum hydrocarbons, such as mineral oil and petrolatum; and water may also be used in suspension formulations.
Suspension can thus include an aqueous liquid or a non-aqueous liquid, an oil-in-water liquid emulsion, or a water-in-oil emulsion.
[219] Dosage forms for oral administration may be aqueous suspensions such as aqueous oral dispersions, emulsions, solutions, and syrups (see, e.g., Singh et al. 2002).
In addition to the active agents, the liquid dosage forms may comprise additives, such as one or more (a) disintegrating agents, (b) dispersing agents, (c) wetting agents, (d) preservatives, (e) viscosity enhancing agents, (f) sweetening agents, and/or (g) flavoring agents. In addition to the additives listed above, the liquid formulations of the invention, in some embodiments, may also comprise inert diluents commonly used in the art such as water or other solvents, solubilizing agents, emulsifiers, flavoring agents, and/or sweeteners. Co-solvents and adjuvants also may be added to a formulation.
[220] In some embodiments, effervescent powders containing the compositions of the invention may be prepared. Effervescent salts are used to disperse medicines in water for oral administration. Effervescent salts also may be packaged as single dose or multi-dose drink mixes, alone or in combination with other ingredients, such as vitamins or electrolytes.
Effervescent salts are granules or coarse powders containing a medicinal agent in a dry mixture, usually composed of sodium bicarbonate and sodium carbonate, citric acid, and/or tartaric acid.
When salts of the invention are added to water, the acids and the base react to liberate carbon dioxide gas, thereby causing "effervescence." Any acid-base combination that results in the liberation of carbon dioxide may be used, as long as the ingredients are suitable for pharmaceutical use, and result in a pH of about 6.0 or higher.
[221] In some embodiments, the compositions of the invention are formulated in a pharmaceutically acceptable transdermal application, and delivered transdermally. Generally speaking, transdermal delivery involves contacting the formulations of the invention with a subject's skin under conditions effective for the active agent(s) to penetrate the skin and cause an effect. Formulations may be prepared as ointments, creams, suspensions, lotions, pastes, gels, sprays, foams, oils, and the like, and any combination thereof.
[222] An exemplary transdermal delivery form is a transdermal "patch" which contains the pharmaceutical compositions. Transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the invention in controlled amounts. Such patches may be constructed for continuous, gradual, pulsatile, or on demand delivery of pharmaceutical agents. A "patch" within the meaning of the invention may be simply a medicated adhesive patch, i.e., a patch impregnated with a composition of the invention for application onto the skin. Thus, a patch may be a single-layer or multi-layer drug-in-adhesive patch, wherein the one or more adhesive layers also contain the active agents. A patch may also be a "matrix" (or -monolithic") patch, wherein the adhesive layer surrounds and overlays the drug layer (wherein a solution or suspension of the active agents is in a semisolid matrix). A
"reservoir" patch may al so be used, comprising a drug layer, typically as a solution or suspension of the active agents in a liquid compartment (i.e., the reservoir), separate from an adhesive layer. A patch also may be part of a delivery system, for instance used with an electronic device communicatively coupled to the mobile device of a user, and coupled with a mobile application (e.g., to control the delivery rate from the reservoir, and optionally to provide information about delivery back to the application or user). Various transdermal patch technologies may be accordingly utilized.
[223] In some embodiments, the compositions of the invention may also be prepared as formulations designed for subcutaneous, intravenous, intra-arterial, intraperitoneal, intraosseous, intramuscular, intrathecal, or intracerebroventricular, injection. Injection formulations may be prepared by dissolving, suspending, or emulsifying the active agent(s) in an aqueous or nonaqueous solvent, non-limiting examples of which include oils, such as vegetable oil, synthetic aliphatic acid glycerides, and esters of higher aliphatic acids or propylene glycol; and may also contain additives such as solubilizers, stabilizers, and suspending, preserving, wetting, emulsifying, dispensing, and isotonic agents.
[224] In some embodiments, the compositions of the invention are formulated in a pharmaceutically acceptable nanostructured formulation, such as a nanoemulsion, a nanocapsule, a nanoparticle conjugate, or a nano-encapsulated oral or nasal spray. Preparations of the compositions of the invention as certain nanostructured formulations may be done by reference to the general knowledge of the art (see, e.g., Jaiswal 2015).
[225] The prefix "nano" as used in the terms describing various embodiments of a nanostructured formulation denotes a size range in the nanometer ("nm") scale.
Accordingly, sizes of such nanoparticle delivery vehicles include those in the range of about 1 to about 100 nm, about 100 to about 200 nm, about 200 to about 400 nm, about 400 to about 600 nm, about 600 to about 800 nm, and about 800 to about 1000 nm, as well as "microparticles" in the range of about 1000 to about 2000 nm (1-2 micrometer ("1.1m") scale). Particles of certain sizes may be particularly advantageous depending on the method of administration, as will be immediately appreciated by one of skill (e.g., for oral liquid emulsion versus for transdermal or topical application). In some embodiments, lipid-based nanoparticles (LBNPs) such as liposomes, solid lipid nanoparticles (SLN), or nanostructured lipid carriers (NLC) are used.
[226] Generally, the compositions of the invention are prepared as pharmaceutically acceptable formulations suitable as oral solid or oral liquid dosage forms, administered sublingually, buccally, topically, rectally, vaginally, ocularly, oticly, nasally, cutaneously, topically, and transdermally; or as intravenous, intra-arterial, intraperitoneal, intraosseous, intramuscular, intrathecal, intracerebroventricular, and subcutaneous injection, wherein such injections comprise physiologically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, liposomes, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
[227] The pharmaceutical composition of the invention therefore may be administered via enteral or parenteral means, wherein enteral means includes, but is not limited to, oral solid and oral liquid dosage forms, sublingual, and buccal administration; and parenteral administration means includes, but is not limited to, intravenous, intra-arterial, intraperitoneal, intraosseous, intramuscular, intrathecal, intracerebroventricular, rectal, vaginal, ocular, nasal, cutaneous, topical, otic, transdermal, and subcutaneous administration; in addition to other equivalent means known to those of skill.
[228] The term "administering" or "administration" broadly refers to providing a compound or pharmaceutical composition of the invention to a subject suffering from, or at risk of, the diseases or conditions to be treated or prevented. As used herein, the terms "subject,- "user,"
"patient," and "individual" are used interchangeably, and refer to any mammal although preferably a human. Such terms will be understood to include one who has an indication for which a compound, composition, or method described herein may be efficacious, or who otherwise may benefit by the invention. In general, all of the compounds, compositions, and methods of the invention will be appreciated to work for all individuals, although individual variation is to be expected, and will be understood.
[229] A "route of administration" is the path by which the compound or composition is taken into the body. Routes of administration may be generally classified by the location at which the substance is applied. Common examples may include oral and intravenous administration.
Routes can also be classified based on where the target of action is. Action may be topical (local), enteral (system-wide effect, but delivered through the gastrointestinal tract), or parenteral (systemic action, but delivered by routes other than the GI tract).
[230] Enteral administration includes administration involving any part of the gastrointestinal tract. The examples may include those by mouth (orally), including oral solid and oral liquid dosage forms, and may be preferably formulated as tablets or capsules.
[231] Parenteral administration refers to administration from any means not involving the gastrointestinal tract, including intravenous (into a vein), intra-arterial (into an artery), intraosseous infusion (into the bone marrow), intramuscular, intracerebral (into the brain parenchyma), intracerebroventricular (into cerebral ventricular system), intrathecal (an injection into the spinal canal), otic (through the ear), ocular (through the eye), rectal, vaginal, and subcutaneous (under the skin). In some embodiments, parenteral administration may include sublingual and/or buccal administration. In some embodiments, a pharmaceutical composition may be administered to a subject via injection. In some embodiments, a pharmaceutical composition may be administered to a subject via nasal systems or the mouth through, for example, an oral solid and/or oral liquid dosage forms;
inhalation, via a nasal spray or oral inhaler; nebulization, from a "nebulizer," which is a small machine that turns liquid medicine into a mist; or buccally/sublingually. In some embodiments, a pharmaceutical composition may be administered to a subject via a combination of administration means. In some embodiments, the pharmaceutical composition may be administered to a subject via one or more enteral administration means. In some embodiments, the pharmaceutical composition may be administered to a subject via one or more parenteral administration means. In some embodiments, the pharmaceutical composition may be administered to a subject via at least one enteral administration means, and at least one parenteral administration means. In some embodiments, an equivalent route of administration known to one of skill is utilized.
[232] By way of non-limiting and merely suggestive example, the following formulations and associated routes of administration may be used in methods of the invention:
EXAMPLE I: Formulation of capsules with 125 ntg or 62.5 mg MDMA
Gelatin capsules, containing the below milligram amounts of MDMA are made as follows:
'Txeitip10), 125 mg _41/U1,4 Capsule ¨
Ingredient Quantity (mg/capsule) MDMA 125.0 Starch 73.0 Magnesium stearate 2.0 ::.41veittplaty 62.5 mg MDMA Capsule Ingredient Quantity (mg/capsule) MDMA 62.5 Starch 36.5 Magnesium stearate 1.0 MDMA is synthesized through known methods or commercially sourced. The MDMA, starch, and magnesium stearate are blended, passed through a No. 20 mesh U.S. sieve, and filled into size 4 hard or soft gelatin capsules in 200 mg or 100 mg quantities as above.
EXAMPLE 2: Formulation of tablets with 125 mg or 62.5 mg MDMA
Tablets, containing the below milligram amounts of MDMA, are made as follows:

Exemplary 125 mg MMIL4 Tablet Ingredient Quantity (mg/tablet) MDMA 125.0 Cellulose, microcrystalline 100.0 Colloidal silicon dioxide 65.0 Stearic acid 10.0 -Evemplary 625 mg AMNIA Tablet Ingredient Quantity (mg/tablet) MDMA 62.5 Cellulose, microcrystalline 50.0 Colloidal silicon dioxide 32.5 Stearic acid 5.0 The components are blended and compressed into tablets, weighing 300 mg or 150 mg total.
[233] It should be readily appreciated that the above formulation examples are illustrative only. Accordingly, any of the compounds may be substituted with the same compound in a different dosage amount (e.g., 120 mg, 115 mg, 110 mg, 105 mg, 100 mg, 95 mg, 90 mg, 85 mg, 80 mg, 75 mg, 70 mg, 65 mg, 60 mg, 55 mg, 50 mg, 45 mg, 40 mg, 35 mg, 30 mg, 25 mg, 20 mg, 15 mg, 10 mg, 5 mg, and values in between). It will be understood that reference to particular compounds is merely illustrative, and both active and inactive compounds in any Example may be substituted by other compounds of the invention. For example, racemic MDMA may be substituted with an enantiomerically enriched composition of MDMA, in an enantiomeric excess in any amount greater than 0%, or a pure or substantially pure individual enantiomer, such as an enantiomer in an enantiomeric excess of greater than about 75%, about 76%, about 77%, about 78%, about 79%, about 80%, about 81%, about 82%, about 83%, about 84%, about 85%, about 86%, about 87%, about 88%, about 89%, or preferably above about 90%, such as about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, about 99.5%, about 99.9%, and up to about 100% and including 100%, as well as values in between.
[234] While in some preferred embodiments, a compound of the invention as used, e.g., in a composition or method of the invention, is MDMA, in other embodiments, a compound will be another therapeutic entactogen. A "therapeutic entactogen- refers to any member of the class of psychoactive compounds that produce distinctive entactogenic effects similar to those of MDMA. Examples of therapeutic entactogens will be known to those in the art, and may for example include substances derived from the 1,3-benzodioxole, cathinone, benzofuran, aminoindane, indole, and amphetamine classes (see, e.g., Oeri 2021).
[235] Accordingly, although in preferred embodiments the therapeutic entactogen is MDMA, in other embodiments a therapeutic entactogen may be, as non-limiting examples, 141,3-benzodioxo1-5-y1)-2-butanamine (BDB); 1-(1,3-benzodioxo1-5-y1)-N-methy1-2-butanamine (MBDB), 3,4-methylenedioxyamphetamine (MDA), 3,4-methylenedioxy-N- ethyl-amphetamine (MDEA). In other embodiments, a therapeutic entactogen may be 3-methylmethcathinone (3-MMC), mephedrone (4-MMC), methylone, a-ethyltryptamine (aET), a-methyltryptamine (aMT), 5-iodo-2-aminoindane (5-IAI), methylene-dioxyaminoindane (MDAI), methylone, ethylone, butylone, 5-(2-Aminopropy1)-2,3- dihydro-1H-indene (5-APDI, IAP), 6-(2-Aminopropyl)benzofuran (6-APB) (b enzofury), 5-(2-aminopropyl)benzofuran (5-APB), 5-MAPB, MDAI, MDOH, or 4-FA (see, e.g., Oeri and works cited). In yet other embodiments, a therapeutic entactogen of the invention is any substituted methylenedioxy- phenethylamine or "MDxx" compound, as will be understood by those in the art. In further embodiments, a therapeutic entactogen is an analog or derivative of an MDxx compound, such as a compound where the methylenedioxyphenyl ring is retained but the phenethyl portion is modified, or a compound which retains the 3,4-cyclised amphetamine core common to the MDxx compounds, but has the 1,3-benzodioxole ring replaced by related heterocycles. In some embodiments, a therapeutic entactogen is a "2C-x"
compound with entactogenic or primarily entactogenic effects.
[236] In some embodiments, a therapeutic entactogen will be any other compound generally known to those of ordinary skill in the art as producing "entactogenic" or "empathogenic"
effects, and which may also be used in "psychedelic--assisted therapy, further including single enantiomers and enantiomeric mixtures; salts and solid forms such as polymorphs, hydrates, solvates, and co-crystals, amorphous forms, deuterated and halogenated versions, and prodrugs, metabolites, analogs, and derivatives of any of the above, including combinations thereof, and further including novel chemical compounds or NCEs having similar structures and/or effects, which may be modified to retain one or more entactogenic effects, but have one or more other novel effects (e.g., minimized side effects, reduced neurotoxicity, optimized onset or course of action, optimized pharmacokinetic or pharmacodynamic properties, and the like).
[237] Moreover, for any of the compounds of the invention, substitution of the compound by its ion, free base, or salt (including in some preferred embodiments, a hydrochloride salt), a polymorph, or an isomer, shall be understood to provide merely an alternative embodiment still within the scope of the invention (with modifications to the formulation and dosage amounts made according to the teachings herein and ordinary skill). Further, compositions within the scope of the invention should be understood to be open-ended and may include additional active or inactive compounds and ingredients. In some embodiments, the composition will contain one or more additional compounds that provide a synergistic effect.
[238] The type of formulation employed for the administration of the compounds employed in the methods of the invention generally are dictated by the compound(s) employed, the type of pharmacokinetic profile desired from the route of administration and the compound(s), and the state of the patient. It will be readily appreciated that any of the above embodiments and classes of embodiments can be combined to form additional embodiments and formulations.
C. Dosage
[239] The invention as disclosed herein provides methods for using therapeutically effective amounts of the pharmaceutical compositions and formulations of the invention in a mammal, and preferably a human. Such methods include those for modulating neurotransmission and for treating a disorder such as a mental health or psychiatric disorder, and specifically AUD.
[240] The pharmaceutical compositions disclosed herein comprise therapeutic amounts of MDMA and/or other active or inactive ingredients. In some embodiments, where a pharmaceutical composition includes MDMA, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 21 mg, 22 mg, 23 mg, 24 mg, 25 mg, 26 mg, 27 mg, 28 mg, 29 mg, 30 mg, 31 mg, 32 mg, 33 mg, 34 mg, 35 mg, 36 mg, 37 mg, 38 mg, 39 mg, 40 mg, 41 mg, 42 mg, 43 mg, 44 mg, 45 mg, 46 mg, 47 mg, 48 mg, 49 mg, 50 mg, 51 mg, 52 mg, 53 mg, 54 mg, 55 mg, 56 mg, 57 mg, 58 mg, 59 mg, 60 mg, 61 mg, 62 mg, 63 mg, 64 mg, 65 mg, 66 mg, 67 mg, 68 mg, 69 mg, 70 mg, 71 mg, 72 mg, 73 mg, 74 mg, 75 mg, 76 mg, 77 mg, 78 mg, 79 mg, 80 mg, 81 mg, 82 mg, 83 mg, 84 mg, 85 mg, 86 mg, 87 mg, 88 mg, 89 mg, 90 mg, 91 mg, 92 mg, 93 mg, 94 mg, 95 mg, 96 mg, 97 mg, 98 mg, 99 mg, 100 mg, 101 mg, 102 mg, 103 mg, 104 mg, 105 mg, 106 mg, 107 mg, 108 mg, 109 mg, 110 mg, 111 mg, 112 mg, 113 mg, 114 mg, 115 mg, 116 mg, 117 mg, 118 mg, 119 mg, 120 mg, 121 mg, 122 mg, 123 mg, 124 mg, 125 mg, 126 mg, 127 mg, 128 mg, 129 mg, 130 mg, 131 mg, 132 mg, 133 mg, 134 mg, 135 mg, 136 mg, 137 mg, 138 mg, 139 mg, 140 mg, 141 mg, 142 mg, 143 mg, 144 mg, 145 mg, 146 mg, 147 mg, 148 mg, 149 mg, 150 mg, 151 mg, 152 mg, 153 mg, 154 mg, 155 mg, 156 mg, 157 mg, 158 mg, 159 mg, 160 mg, 161 mg, 162 mg, 163 mg, 164 mg, 165 mg, 166 mg, 167 mg, 168 mg, 169 mg, 170 mg, 171 mg, 172 mg, 173 mg, 174 mg, 175 mg, 176 mg, 177 mg, 178 mg, 179 mg, 180 mg, 181 mg, 182 mg, 183 mg, 184 mg, 185 mg, 186 mg, 187 mg, 188 mg, 189 mg, 190 mg, 191 mg, 192 mg, 193 mg, 194 mg, 195 mg, 196 mg, 197 mg, 198 mg, 199 mg, 200 mg, and values in between.
[241] In some embodiments, a patient may be administered an initial dose of between 1 mg to 200 mg of MDMA, including 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 21 mg, 22 mg, 23 mg, 24 mg, 25 mg, 26 mg, 27 mg, 28 mg, 29 mg, 30 mg, 31 mg, 32 mg, 33 mg, 34 mg, 35 mg, 36 mg, 37 mg, 38 mg, 39 mg, 40 mg, 41 mg, 42 mg, 43 mg, 44 mg, 45 mg, 46 mg, 47 mg, 48 mg, 49 mg, 50 mg, 51 mg, 52 mg, 53 mg, 54 mg, 55 mg, 56 mg, 57 mg, 58 mg, 59 mg, 60 mg, 61 mg, 62 mg, 63 mg, 64 mg, 65 mg, 66 mg, 67 mg, 68 mg, 69 mg, 70 mg, 71 mg, 72 mg, 73 mg, 74 mg, 75 mg, 76 mg, 77 mg, 78 mg, 79 mg, 80 mg, 81 mg, 82 mg, 83 mg, 84 mg, 85 mg, 86 mg, 87 mg, 88 mg, 89 mg, 90 mg, 91 mg, 92 mg, 93 mg, 94 mg, 95 mg, 96 mg, 97 mg, 98 mg, 99 mg, 100 mg, 101 mg, 102 mg, 103 mg, 104 mg, 105 mg, 106 mg, 107 mg, 108 mg, 109 mg, 110 mg, 111 mg, 112 mg, 113 mg, 114 mg, 115 mg, 116 mg, 117 mg, 118 mg, 119 mg, 120 mg, 121 mg, 122 mg, 123 mg, 124 mg, 125 mg, 126 mg, 127 mg, 128 mg, 129 mg, 130 mg, 131 mg, 132 mg, 133 mg, 134 mg, 135 mg, 136 mg, 137 mg, 138 mg, 139 mg, 140 mg, 141 mg, 142 mg, 143 mg, 144 mg, 145 mg, 146 mg, 147 mg, 148 mg, 149 mg, 150 mg, 151 mg, 152 mg, 153 mg, 154 mg, 155 mg, 156 mg, 157 mg, 158 mg, 159 mg, 160 mg, 161 mg, 162 mg, 163 mg, 164 mg, 165 mg, 166 mg, 167 mg, 168 mg, 169 mg, 170 mg, 171 mg, 172 mg, 173 mg, 174 mg, 175 mg, 176 mg, 177 mg, 178 mg, 179 mg, 180 mg, 181 mg, 182 mg, 183 mg, 184 mg, 185 mg, 186 mg, 187 mg, 188 mg, 189 mg, 190 mg, 191 mg, 192 mg, 193 mg, 194 mg, 195 mg, 196 mg, 197 mg, 198 mg, 199 mg, 200 mg, and values in between, and then subsequently administered a booster dose of between 0.5 mg to 100 mg of MDMA, including 0.5 mg, 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 21 mg, 22 mg, 23 mg, 24 mg, 25 mg, 26 mg, 27 mg, 28 mg, 29 mg, 30 mg, 31 mg, 32 mg, 33 mg, 34 mg, 35 mg, 36 mg, 37 mg, 38 mg, 39 mg, 40 mg, 41 mg, 42 mg, 43 mg, 44 mg, 45 mg, 46 mg, 47 mg, 48 mg, 49 mg, 50 mg, 51 mg, 52 mg, 53 mg, 54 mg, 55 mg, 56 mg, 57 mg, 58 mg, 59 mg, 60 mg, 61 mg, 62 mg, 63 mg, 64 mg, 65 mg, 66 mg, 67 mg, 68 mg, 69 mg, 70 mg, 71 mg, 72 mg, 73 mg, 74 mg, 75 mg, 76 mg, 77 mg, 78 mg, 79 mg, 80 mg, 81 mg, 82 mg, 83 mg, 84 mg, 85 mg, 86 mg, 87 mg, 87, 88 mg, 89 mg, 90 mg, 91 mg, 92 mg, 93 mg, 94 mg, 95 mg, 96 mg, 97 mg, 98 mg, 99 mg, 100 mg, and values in between. In some embodiments, the booster dose of MDMA will be 1/2 of the initial dose of MDMA, such that, in an exemplary embodiment, if a subj ect were administered 100 mg of MDMA in an initial dose, the second such dose administered to the subject would be 50 mg. In some embodiments, such as in certain aspects of Examples 4 or 8, wherein a subtherapeutic or "active" control of 20 or 10 mg is used, capsules may be prepared as in Example 1 (or tablets as in Example 2), but with amounts of the active and inactive ingredients adjusted accordingly. In some embodiments, the booster dose of MDMA is greater than 100 mg, such as 125 mg, 150 mg, and 175 mg, and including up to 200 mg.
[242] In some embodiments, a subject is only administered a single dose of MDMA, wherein the single dose of MDMA is between 1 mg to 200 mg of MDMA, including 1 mg to 200 mg of MDMA, including 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 21 mg, 22 mg, 23 mg, 24 mg, 25 mg, 26 mg, 27 mg, 28 mg, 29 mg, 30 mg, 31 mg, 32 mg, 33 mg, 34 mg, 35 mg, 36 mg, 37 mg, 38 mg, 39 mg, 40 mg, 41 mg, 42 mg, 43 mg, 44 mg, 45 mg, 46 mg, 47 mg, 48 mg, 49 mg, 50 mg, 51 mg, 52 mg, 53 mg, 54 mg, 55 mg, 56 mg, 57 mg, 58 mg, 59 mg, 60 mg, 61 mg, 62 mg, 63 mg, 64 mg, 65 mg, 66 mg, 67 mg, 68 mg, 69 mg, 70 mg, 71 mg, 72 mg, 73 mg, 74 mg, 75 mg, 76 mg, 77 mg, 78 mg, 79 mg, 80 mg, 81 mg, 82 mg, 83 mg, 84 mg, 85 mg, 86 mg, 87 mg, 88 mg, 89 mg, 90 mg, 91 mg, 92 mg, 93 mg, 94 mg, 95 mg, 96 mg, 97 mg, 98 mg, 99 mg, 100 mg, 101 mg, 102 mg, 103 mg, 104 mg, 105 mg, 106 mg, 107 mg, 108 mg, 109 mg, 110 mg, 111 mg, 112 mg, 113 mg, 114 mg, 115 mg, 116 mg, 117 mg, 118 mg, 119 mg, 120 mg, 121 mg, 122 mg, 123 mg, 124 mg, 125 mg, 126 mg, 127 mg, 128 mg, 129 mg, 130 mg, 131 mg, 132 mg, 133 mg, 134 mg, 135 mg, 136 mg, 137 mg, 138 mg, 139 mg, 140 mg, 141 mg, 142 mg, 143 mg, 144 mg, 145 mg, 146 mg, 147 mg, 148 mg, 149 mg, 150 mg, 151 mg, 152 mg, 153 mg, 154 mg, 155 mg, 156 mg, 157 mg, 158 mg, 159 mg, 160 mg, 161 mg, 162 mg, 163 mg, 164 mg, 165 mg, 166 mg, 167 mg, 168 mg, 169 mg, 170 mg, 171 mg, 172 mg, 173 mg, 174 mg, 175 mg, 176 mg, 177 mg, 178 mg, 179 mg, 180 mg, 181 mg, 182 mg, 183 mg, 184 mg, 185 mg, 186 mg, 187 mg, 188 mg, 189 mg, 190 mg, 191 mg, 192 mg, 193 mg, 194 mg, 195 mg, 196 mg, 197 mg, 198 mg, 199 mg, 200 mg, and values in between.
[243] In some embodiments, a single dose of MDMA will be between 25 mg and 175 mg, preferably between 50 mg and 150 mg, and more preferably between 62.5 mg and 125 mg, inclusive. In one embodiment, a MDMA-assisted psychotherapy regime of two doses of MDMA is used, with an "initial dose" of 125 mg and a "booster dose" two hours later of 62.5 mg. In another embodiment, a MDMA-assisted psychotherapy regime of two doses of MDMA
is used, with an initial dose of 100 mg and a "booster" dose two hours later of 50 mg. In another embodiment, a MDMA-assisted psychotherapy regime of two doses of MDMA is used, with an initial dose of 80 mg and a "booster" dose two hours later of 40 mg. In some embodiments where two doses are administered to a patient, the booster dose will be 0.5x (1/2) or about 0.5x (1/2) of the initial dose (see, e.g., Sessa et al. 2021). In some embodiments, the booster dose will be chosen based on objective or subjective patient measures, based on patient selection or choice at or preceding the time of administration, or based on the exercise of clinical judgment at or preceding the time of administration. In some embodiments, participants may receive a third or further dose (i.e., a second or further booster dose), which may be the same, a greater, or a lesser dose than the first booster dose.
[244] Doses of MDMA ranging from 100-125 mg reach peak plasma concentration in humans after 2-3 hours. In patients with PTSD administered this dosing regime, onset of action occurred 45-75 minutes after the initial dose and peak drug effects were reported at 2-5 hours (Mithoefer et al. 2013). For those receiving one dose, the effects lasted 4-5 hours, which was extended with the booster dose to 5-6 hours. In a prior study, subjective effects were reported after approximately 45 mins, peaking around 1.5-2 hours, and decreasing to baseline values 5-6 hours later (Harris 2002). The elimination half-life of MDMA is between 7-9 hours, with similar results for doses ranging from 50-125 mg (Mas et al 1999). In brain imaging studies, peak subjective effects have been reported at 100 mins and have lasted approximately 4.5 hours after 100 mg MDMA in healthy volunteers (Carhart-Harris 2013).
[245] It will be readily appreciated that dosages may vary depending upon the treatment protocol itself, the onset, progression, severity, frequency, duration, probability of, or susceptibility of the symptom to which treatment is directed, clinical endpoint desired, previous, simultaneous or subsequent treatments, general health, age, gender, and race of the subject, bioavailability, potential adverse systemic, regional, or local side effects, the presence of other disorders or diseases in the subject, and other factors that will be appreciated by the skilled artisan (e.g., medical or familial history). For example, the upper age range of subjects may be limited, e.g., to 65 years, and patients may be screened for cardiac abnormalities, and for medications that may affect the metabolism of MDMA (e.g., through CYP2D6 or CYP3A4), or genetic variations that may affect metabolism of MDMA.
[246] Dose amount, frequency or duration may be increased or reduced, as indicated by the clinical outcome desired, status of the pathology or symptom, any adverse side effects of the treatment or therapy, or concomitant medications. For example, as MDMA at doses of 100-125 mg has been associated with increases in heart rate (of 26-30 bpm) and blood pressure (of diastolic 14.4-25 mmg) (Harris 2002, Mas 1999, Mithoefer et al. 2011) these may be carefully monitored following the regime disclosed elsewhere (Oehen et al. 2013, Chabrol & Oehen 2013), and patients experiencing particular symptoms may not receive a booster dose. The skilled artisan with the teaching of this disclosure in hand will appreciate the factors that may influence the dosage, frequency, and timing required to provide an amount sufficient or effective for providing a therapeutic effect or benefit, and to avoid or minimize adverse effects.
[247] It will be understood that, in some embodiments, the dose actually administered will be determined by a physician, in light of the relevant circumstances, the method of delivery, (i.e., methods that are systemic and that are not subject to first pass effect will require less of a dose than those which are metabolised prior to entering the bloodstream; the age of the patient, the weight of the patient, whether the patient has any coin orbi diti es (i.e., any other medical conditions simultaneously present within a patient), other medications the patient is taking (routinely or presently), and any patient-specific aspects that could affect the way in which the pharmaceutical composition interacts with the patient, such as variations in metabolism, variations in patient response, etc., and therefore any dosage ranges disclosed herein are not intended to limit the scope of the invention. In some instances, dosage levels below the lower limit of a disclosed range may be more than adequate, while in other cases doses above a range may be employed without causing any harmful side effects, provided for instance that such larger doses also may be divided into several smaller doses for administration, either taken together or separately.
[248] In such embodiments, the pharmaceutical compositions may be administered and dosed in accordance with good medical practice, taking into account the method and scheduling of administration, prior and concomitant medications and medical supplements, the clinical condition of the individual patient and the severity of the underlying disease, the patient's age, sex, body weight, and other such factors relevant to medical practitioners, and knowledge of the particular compound(s) used. Dosage levels thus may differ from patient to patient, for individual patients across time, and for different pharmaceutical compositions and formulations, but shall be able to be determined with ordinary skill.
Determination of appropriate dosing shall include not only the determination of single dosage amounts, but also the determination of the number and timing of doses, and the time(s) of day or time(s) during a psychotherapeutic session preferable for their administration.
[249] In some embodiments, especially where a formulation is prepared in single unit dosage form, suggested dosage amounts shall be known by reference to the format of the preparation itself. In other embodiments, suggested dosage amounts may be known by reference to the means of administration or by reference to the packaging and labeling, package insert(s), marketing materials, training materials, or other information and knowledge available to those of skill or the public. Another aspect of this disclosure therefore provides pharmaceutical kits containing a pharmaceutical composition or formulation of the invention, suggested administration guidelines or prescribing information therefor, and a suitable container.
Individual unit dosage forms can be included in multi-dose kits or containers.
Pharmaceutical formulations also can be packaged in single or multiple unit dosage forms for uniformity of dosage and ease of administration.
D. Methods of Treating AUD and Administering the Composition
[250] The pharmaceutical compositions disclosed herein are shown to be useful in methods for treating AUD, as in the exemplary treatment methods now described.
[251] In some embodiments, prior to beginning a therapy protocol, a patient will be enrolled in a preparation group. In such embodiments, a person is first assessed to determine whether they are eligible for treatment at screening (i.e., whether they meet all inclusion criteria, as defined herein, and do not meet any exclusion criteria, as defined herein). If so, the person may participate in the preparation group.
[252] In some embodiments, the preparation group will span four weeks and include content such as "building confidence" which includes discussing problems drinking has caused in the patient's family, job, social life, health, etc.; why change is necessary (completing a cost benefit analysis worksheet), discussing when something went well for the patient, completing a drinking diary, where drinking is monitored daily for one week; and completing "homework"

tasks, such as the drinking diary and a worksheet detailing the patient's strengths and weaknesses; "resources, barriers, and goal setting" which includes discussing goal setting as it relates to drinking, reviewing potential barriers and resources that may aid in the reduction of alcohol consumption, such as discussing the patient's strengths and qualities, and identifying social and community support outlets, completing homework tasks, such as setting a goal related to alcohol consumption, completing a daily diary, and completing a worksheet that details social and community support outlets; "increasing rewarding activities and building support" which includes identifying challenges that may hinder the accomplishment of goals, and discussing how those challenges were/would be managed; reviewing the social and community support worksheet, and working as a group to build upon the resources listed within; setting rewarding goals, including planning pleasant activities, etc.;
completing homework tasks, which may include setting a goal related to drinking, and beginning a pleasant activities planner; and "building the foundations for recovery" which includes discussing how the patient will manage moving forward, including identifying anticipated challenges and brainstorming ways to manage those, identifying any changes the patient would like to make in their recovery program, and identifying such changes if necessary;
completing a recovery support map, and sharing/discussing it with someone who supports the patient In other embodiments, a preparation group may contain more or less content such as the above, and/or be for a shorter or longer period of time
[253] In some embodiments, if a patient successfully completes the preparation group course (wherein "completes" in this context may refer to, e.g., the patient successfully reducing their alcohol intake as compared to their alcohol intake prior to the start of the preparation group course and/or the patient actively participating in the preparation group, such that the patient readily collaborates with the other members of the preparation group and fully or substantially completes each task required in the preparation group), and the patient remains eligible for treatment (e.g., meets all required "inclusion criteria" and does not meet at least one "exclusion criteria,") the patient may begin a treatment protocol.
[254] In some embodiments, the methods of the invention may reduce relapse rates (wherein "relapse- refers to a resumption of drinking at a level that is above the threshold of "harmful use" as quantified by the ICD-10). In some embodiments, the methods of the invention may reduce relapse rates by increasing the participant's "psychological flexibility," wherein psychological flexibility refers to a person's ability to be responsive and adaptive to situations as they arise, to shift perspectives (as opposed to being rigid or stuck') and to act mindfully in accordance with core values (rather than reacting impulsively or on 'auto-pilot').
[255] In some embodiments, the methods of the invention will include therapy sessions employing acceptance and commitment therapy (ACT), an enhanced form of cognitive behavioral therapy (CBT), which focuses upon an individual's relationship with their internal experiences (e g , thoughts, emotions, physical sensations, memories) rather than the content of these experiences, and uses mindfulness as one of the cornerstones from which to explore this The main purpose of ACT is to increase psychological flexibility via six core processes of being mindfully present, stepping back and being willing to accept all internal experiences, defusing thoughts, taking an observer's perspective, awareness of values (e.g., what's important), and acting in accordance with those values.
[256] In some embodiments, the methods of the invention are useful in helping an individual achieve a more fulfilling and meaningful life (via increased psychological flexibility). A
reduction in the levels of distress are typically a welcome by-product of ACT
interventions (Harris, 2009). ACT can be seen as supporting and building upon the core mechanism of psychedelic-assisted therapies which enhance psychological flexibility, alongside enhancing the relapse prevention objective in supporting people to achieve a more fulfilling life without alcohol.
[257] Relapse prevention can be an integral part of alcohol treatment. This intervention typically involves supporting individuals to develop greater insight regarding the potential triggers and high risk situations for using alcohol, identifying early warning signs and, crucially, developing effective coping skills and enhanced self-efficacy to manage these situations (Hendershot et al., 2011). Mindfulness-based relapse prevention (Marlatt et al., 2011) incorporates core elements of mindfulness skills into this intervention.
Mindfulness can be defined as "paying attention in a particular way: on purpose, in the present moment, and non-judgmentally" (Kabat-Zinn, 1994) and has been embedded across a range of therapeutic interventions including CB T for depression (Teasdale et al., 2000) and stress-reduction (Kabat-Zinn et al., 1992). In relation to relapse prevention, mindfulness-based principles can be incorporated via helping individuals to pay attention to their internal experiences (e.g., thoughts and emotions) and their potential role in relapse and practice in incorporating mindfulness practices into daily life (Bowen et al,, 2009).
[258] In some embodiments, the methods of the invention employ one or more elements of mindfulness skills, for example in mindfulness-based relapse prevention, or otherwise employ mindfulness-based principles or mindfulness practices, including for purposes of relapse prevention, or to provide or support one or more other therapeutic effects, including: (a) reducing alcohol use, (b) reducing alcohol cravings, (c) promoting alcohol abstinence, (d) preventing relapse into alcohol use, or (e) improving at least one symptom of alcohol use disorder. In some embodiments, the one or more elements of mindfulness skills, or employment of mindfulness-based principles or mindfulness practices will provide or support one or more of: (a) increasing quality of life, (b) increasing psychosocial functioning, (c) decreasing use or frequency of a prescription medication, (d) decreasing use or frequency of a recreational drug, (e) decreasing obsessive compulsive thoughts, (f) decreasing suicidality, (g) increasing feelings of empathy, (h) increasing self-compassion, or (i) improving at least one symptom of a comorbid psychiatric disorder.
[259] In some embodiments, the methods of the invention employ motivational interviewing (MI), a technique used to enhance motivation to change behavior that is widely used in addiction services. Miller & Rollnick (1991) describe a key element of this process as coming alongside the individual to explore ambivalence and elicit the person's own ideas about change.
MI is characterized by therapists' unconditional acceptance of where the individual finds themselves and their perspective. In MI, a therapist's role is to work in partnership and collaboration (not to act as the expert, to lecture or argue), to be genuinely curious and ask questions that provide opportunities for the individual to hear themselves talk about change and to have compassion for the individual (to prioritise their needs and an authentic regard for their well-being).
EXAMPLE 3: Exemplary Treatment Protocols¨"Harmful Use"
[260] In certain exemplary treatment methods wherein the compositions of the invention are shown to be useful in methods for treating non-physically dependent AUD, the treatment flow can be conceived in phases as follows, and are specifically outlined in FIG.
12: screening and initial eligibility check (1201); pre-MDMA course preparatory group therapy course, e.g., a four-week course of weekly sessions (1202); Baseline visit and eligibility confirmation (1203);
an MDMA-assisted psychotherapy course, e.g., over eight or 13 weeks (1204);
follow-up and data collection, e.g., at 3 months, 6 months, and 9 months (1205).
[261] It will be readily appreciated, however, that this exemplary treatment flow is merely illustrative of one use of the pharmaceutical compositions of the invention in a method of treating AUD, that a use need not include every illustrated aspect (e.g., the follow-up or data collection of step six), or be in the exact order below, and moreover, that the steps, stages, or phases below (and their delineation as such) are simply for purposes of assisting the reader with the conceptualization of a use of the invention, whereas the full breadth and scope of the invention shall be known by reference to the appended claims.
TABLE 4: Exemplary Treatment Flow¨"Iarmfill Use"
1. Screening and Eligibility Check 2. Pre-MDMA Course Preparatory Group Therapy Course 3. Baseline Assessment Visit and Eligibility Confirmation 4. MDMA-Assisted Psychotherapy Course 5. Follow-Up and Data Collection Screening and Initial Eligibility Check (101)
[262] Patients who meet the diagnostic criteria for AUD, but are not physically dependent, meaning detoxification is not required, will be screened prior to entering the pre-MDMA
course group therapy alcohol reduction program. Eligible patients will attend a baseline visit after the pre-course group program to ensure eligibility.
[263] Potential patients will be invited to a screening visit where their eligibility for MD1VIA-assisted psychotherapy using the methods of the invention will be determined.
The screening visit will include informed consent, medical, psychiatric and substance use history, prescribed medication use, basic physiological observations and alcohol breath test, urine drugs, and pregnancy screen as clinically indicated. In order to monitor the risk of patients using MDMA
outside of the therapeutic context, they can be asked about their use of, or desire to use, illicit ecstasy, e.g., with standard questions such as those used by MAPS in MDMA-assisted therapy studies.
[264] During screening the therapeutic process will be discussed and explained. The patients' understanding of the procedure, requirements, and commitments will be confirmed and any questions answered before informed consent is obtained. If video and/or audio recordings will be made during therapy sessions, patients will be asked to consent. If patients do not wish to have sessions recorded (or at any point decide to withdraw consent for the whole or part of the session) recordings will be stopped and deleted if requested. Potential patients also may be contacted and partake in a short telephone screen before invitation to a full screening visit, to minimize inconvenience to "obviously" ineligible patients.
[265] In some embodiments, a psychiatric assessment is sought. In such embodiments, it will be performed by a trained psychiatrist Assessments may include, e g (1) the Mini International Neuropsychiatric Interview 5 (MINI 5) (Sheehan et al. 1998) to screen for comorbid psychiatric disorders; (2) diagnostic evaluation of the presence of Alcohol Use Disorder according to DSM-5 (assessed using SCID-5-CT (Clinical Trials Version) (First et al. 2015)); (3) diagnostic evaluation of the presence of AUD in the absence of physical dependence, as guided by the SADQ, and (4) clinical risk assessment for suicidality using the Columbia Suicide Severity Rating Scale (C-SSRS) (Mundt 2013). Clinical data collection may include years of alcohol use and any dependence, complications/consequences, and previous therapeutic inputs provided including any previous in-patient or community alcohol detox programs. Other questionnaires may also be completed, as will be appreciated.
[266] In some embodiments, information about alcohol consumption for the three months previous to the screening visit is assessed using the Alcohol Timeline Follow Back (TLFB) method, the gold-standard in self-report substance use, which utilizes a calendar method and memory aids to collect retrospective estimates of daily drinking to obtain accurate daily estimates of the quantity of alcohol consumption (S ob ell 2001). Patients may receive the same calendar to take away to track their drinking behavior for the duration of the therapy.
Pre-MDMA Course Preparatory Group Therapy Course (102)
[267] In some embodiments, after screening, participants will attend weekly sessions of group therapy, which will be run by a trained professional with expertise in addiction treatment. In some embodiments, participants will attend weekly sessions of group therapy for four weeks, although shorter or longer courses may also be used, and in some embodiments the preparatory group therapy course may be skipped entirely. The purpose of the group will be to provide psychosocial support to assist individuals to gradually reduce their alcohol intake prior to starting the course of MDMA-assisted therapy.
[268] A reduction in daily or weekly alcohol consumption should form part of a structured, individualised care plan involving careful preparation (NICE Guidelines).
Adequate preparation also benefits from consideration of a person's support network, motivation and after-care planning to improve post-treatment outcomes (Kouimtsidis, 2017).
Prior to being considered ready for the MDMA-assisted course, patients thus may attend weekly sessions of group therapy. In some exemplary embodiments, groups of between 5 and 10 participants can be facilitated by one or two trained staff with experience in delivering motivational interview style alcohol treatment group counselling. However, as would be apparent to one of skill, the number of participants and instructors may vary and, in some embodiments, the number of participants may be greater than 10, including at least 12, at least 14, at least 16, at least 18, and at least 20. Likewise, the number of participants may be less than 5, including 4, 3, 2, or 1.
The number of trained staff with experience in delivering motivational interview-style alcohol treatment group counseling may, in some embodiments, be greater than 2, including 3, 4, 5, 6, 7, 8, 9, 10, or more than 10.
[269] In some embodiments, rather than participating in an in-person group therapy course, participants attend a synchronous virtual group therapy course (i.e., participants attend all sessions "live," but remote, e.g., via computer). In some embodiments, participants attend an asynchronous virtual group therapy course (i.e., attend at selected times of their choosing, where less than all participants may attend at the same time), and wherein one or more portions of the course will be pre-recorded. Participants attending a detoxification course (as discussed further below) may also attend a synchronous or asynchronous virtual course.
[270] In some embodiments, each weekly group therapy session will last for between about 30 minutes to about 120 minutes. In some embodiments, weekly group therapy sessions will last for about 30 minutes, about 35 minutes, about 40 minutes, about 45 minutes, about 50 minutes, about 55 minutes, about 60 minutes, about 65 minutes, about 70 minutes, about 75 minutes, about 80 minutes, about 85 minutes, about 90 minutes, about 95 minutes, about 100 minutes, about 105 minutes, about 110 minutes, about 115 minutes, about 120 minutes, and values in between. In some embodiments, weekly group therapy sessions may last for greater than 120 minutes, or may take place in a residential (or "rehab") facility. In some embodiments, groups are closed and confidential. In some embodiments, patients are enrolled into the groups on a rolling basis and will leave after a minimum (e.g., of four) weeks attendance to be enrolled into the MDMA-assisted psychotherapy course when deemed appropriate by the facilitators.
[271] In some embodiments, if a patient is not deemed sufficiently engaged in the preparation phase (e.g., as deemed by the group facilitators) and/or has not managed to reduce their use of alcohol to zero, their enrollment into the MDMA-assisted psychotherapy course can be delayed until they are considered to be adequately prepared or otherwise ready to progress. In some embodiments, wherein further preparation is required, it may include attending further group sessions which, in some embodiments, requires repeating the entire course while, in other embodiments, merely repeating one or more weeks of the course.
Baseline Assessment Visit and Eligibility Confirmation (103)
[272] After a participant has successfully completed the pre-treatment preparation group therapy course, the participant will be assessed for eligibility to start the MDMA-assisted psychotherapy course. This is the "baseline" point. During this visit, continued consent is confirmed and vital signs reassessed (pulse, blood pressure, etc.). A brief physical examination and ECG is performed, and blood samples for routine laboratory tests (urea and electrolytes, full blood count, liver function tests and Gamma-GT) are also completed. A
blood test is not required if the patient has had one in, e.g., the last three months.
MDMA-Assisted Psychotherapy Course ________________ Two MDMA Sessions
[273] In an exemplary embodiment of a MDMA-assisted psychotherapy treatment regimen (an "MDMA-assisted psychotherapy course" or "MDMA-assisted psychotherapy regimen"), a patient will receive ten total therapy sessions during the treatment phase:
eight 60-minute psychotherapy sessions, and two 6-8 hour MDMA-assisted psychotherapy sessions.
Sessions will take place approximately 1-2 weeks apart and start as soon as possible after baseline.
TABLE 5 Exemplary Eight-Week MDMA.-Assisted Pchotherapy Course Week Session Activity 1 1 Non-Drug Prep Session (60-minute therapy session) 2 2 Non-Drug Prep Session (60-minute therapy session) 3 MDMA-Assisted Therapy Session 1 (-6-8 hours) 4 Next-Day Non-Drug Integration / Follow-up Session (60 mins) 4 5 Non-Drug Integration Session (60-minute therapy session) 6 Non-Drug Integration Session (60-minute therapy session) 7 MDMA-Assisted Therapy Session 2 (-6-8 hours) 8 Next-Day Non-Drug Integration / Follow-up Session (60 mins) 7 9 Non-Drug Integration Session (60-minute therapy session) 8 10 Non-Drug Integration & Final Evaluation Session (-90 mins)
[274] MDMA-assisted psychotherapy sessions may be run by a therapist pair, e.g., one male and one female, although they also may be run by two male or two female therapists, one male and one non-binary therapist, one female and one non-binary therapist, two nonbinary therapists, a single therapist, or more than two therapists. In some embodiments, psychological support during each MDMA session preferably will be provided by two members comprising a "therapy team." Where "therapy team" is used herein, it will be appreciated that modifications can be made to accommodate more than two therapists, such as 3, 4, or 5 therapists; or use a single therapist, and in such embodiments "team" will refer to that therapist alone (or where appropriate in context, to that therapist and another therapist or clinician not necessarily present with the patient during an MDMA-assisted or other therapy session, but nevertheless a member of the patient's overall care team). A therapy team, for example, may comprise a psychiatrist and a co-therapist who is a clinical psychologist or psychotherapist, but either member may be a "therapist" as the term is defined above.
[275] A dyadic male-female pair is used in some embodiments herein. A dyadic male-female pair is a feature of certain MDMA-assisted therapy (Greer and Tolbert 1998);
however, the co-therapist pair herein need not necessarily be male-female; same-sex pairs, and pairs comprising at least one member not identifying as a male or a female are also appropriate. Where a dyadic therapy pair is used, in some embodiments, a patient will get to know both therapists in the preparatory sessions before their first MDMA session. In such embodiments, a facilitative relationship is thereby developed, which will aim to reduce discomfort or difficulties that might arise during the course of therapy.
[276] Guidelines for each therapeutic session in exemplary embodiments follow.
In some embodiments, the patient will drive the psychotherapy sessions according to the patient's individual needs. In some embodiments, the therapists guide the psychotherapy sessions. In some embodiments, the therapeutic approach may be guided in part with reference to the Manual for MDMA-Assisted Psychotherapy in the Treatment of PTSD (Mithoefer, 2017)
[277] In some embodiments, at the start of all psychotherapy sessions, patients are asked about alcohol use and complete an alcohol breath test to confirm sobriety during therapy. On all MDMA-assisted psychotherapy sessions, an alcohol breath test and urinalysis (dipstick) for recreational drugs and pregnancy (if applicable) may be performed. A positive result for recreational drugs other than cannabis (THC) on a test would terminate the session.
[278] What now follows is an exemplary session flow, as may be used in some embodiments of the invention; changes, modifications, and additions may be made according to the teachings herein and the general knowledge in the art, and will be appreciated.
[279] Sessions 1 and 2: Therapists will discuss MDMA, how it works, the expected effects, risks and review of the historical evidence of MDMA as a tool for psychotherapy. In some embodiments, the patient will be invited to share aspects of their life history that they feel important for the therapists to know, given that, without being bound by theory, thoughts, feelings, images and memories about past experiences may come to the patient's mind during the MDMA session. Discussion, in some embodiments, will also include questions about the patient's internal experiences (e.g., thoughts, emotions, imagery) and their understanding of this. In such embodiments, the patient will be invited to discuss their current coping responses and any goals/hopes for participating in the study, alongside any intentions for what they would like to explore during the MDMA session. That said, such embodiments are merely exemplary, and should not be construed as limiting the invention as disclosed herein.
Necessarily, in some embodiments, other such questions may be asked by one or more therapists to assess, among other things, the background, needs, and goals of the patient and, likewise, the structure of the psychotherapy session may be modified in response to the way in which the patient is responding, as would be apparent to one of skill.
[280] Session 3 (MDMA-assisted session): Patients will arrive at an agreed time which, in some embodiments, is in the morning¨for instance between approximately 8-9 am.
However, the scheduled time need not be in the morning, and may instead be in the afternoon, the evening, or late at night. Regardless, continued consent will be registered and eligibility may be checked, including assessment of physical health, alcohol breath test and urine drugs screen for recreational drugs and pregnancy (if applicable). Vital sign measurements such as heart rate, blood pressure, and temperature may be taken at baseline before the MDMA is administered and before discharge (or more frequently if clinically indicated, including by use of medical sensors or biosensors that provide continuous, near-continuous, or periodic measurements).
Consent to video and audio record the session may be obtained or confirmed; if it is withdrawn, recording should not take place (or only audio recording should take place, if such specific and limited consent is obtained).
[281] In some embodiments, patients will receive an initial dose of about 125 mg MDMA
(e.g., two capsules or tablets of 62.5 mg each), as in the formulations of Examples 1 and 2 or the equivalent. In other embodiments, patients may receive an initial dose of another amount, e.g., 100 mg or 80 mg. Patients will be invited to relax for about 90 minutes, lying down with eyeshades on if happy to do so. The patient may have the option to listen to music, either through headphones or into the room, and may continue to do so throughout the therapy session. During this initial phase of the effects of MDMA most patients prefer to have little communication but rather to acclimatize to the psychological and physical effects of the drug.
[282] After 90 minutes the patient may be invited to sit and the facilitators may gently guide the patient into discussion of their particular individual aspects of their history.
[283] At an appropriate time after receiving the initial dose of MDMA the patient may be given the option to receive a further supplemental dose of, in some embodiments, half the original dose (i.e., 62.5 mg MDMA, in this example; or, e.g., 50 mg or 40 mg).
In an embodiment, the time of administration of the supplemental dose will be two hours following the administration of the initial dose. In some embodiments, the time of administration of the booster dose will be determined so as to extend peak subjective effects. In some embodiments, the time of administration of the booster dose will be determined so as to optimize the plasma concentration-time profile, e.g., by prolonging the time at higher plasma concentrations. rt he purpose of this is to prolong the psychological and direct pharmacological effects of the drug to allow for further time for psychotherapy, and therefore such prolongation can be optimized or determined either by reference to the subjective (psychological) effects, or the pharmacokinetic (pharmacological) effects, or through other known methods in the art.
[284] Generally, patients will not be encouraged to get up and move around during the session, but rather to remain sitting or lying throughout. However, if a patient wishes to get up, this can be facilitated if the therapy team judges it safe to do so. In some embodiments, physical touch from the therapists toward the patient is employed as a therapeutic tool. In such embodiments, this technique can be used as a supportive grounding technique and to allow some patients to externalize their psychological pain. This technique should be carefully managed with professional boundaries by the therapists and prior informed consent.
[285] The patient may be required to remain in the facility until they are deemed fit to return home. They may, e.g., be assessed by the attendant doctor and signed off as medically fit for discharge. If not deemed ready to leave they may be required to remain in the therapy facility until deemed medically fit for discharge. The patient may then be met at the facility by their pre-arranged significant other, who can accompany them home. Generally, for safety reasons, a patient should not be permitted to leave on their own.
[286] Session 4: The day following session 3, the patient may undertake a follow-up session with the therapy team. This generally will involve a 60-minute face-to-face follow-up session discussing aspects of MDMA-assisted psychotherapy undertaken the day before Patients will be given the contact details of a clinical member of the therapy team whom they can contact if required for further telephone support during the week after each MDMA
session. In some embodiments, patients will receive one or more remote or virtual follow-ups after session 4, for example daily phone calls or using another follow-up means as described further below.
[287] Sessions 5 and 6: The patient may have the opportunity to reflect upon the MDMA
session and to explore the material that emerged. There are opportunities to reflect on new insights or perspectives gained and how this may impact on the issues unpinning their addictive behaviors and/or other areas of difficulty. Intentions for the next MDMA
session may be discussed.
[288] Session 7 (MDMA -assisted session): Same as session 3 above; outlined in TABLE 5.
[289] Session 8: Same as session 4 above; outlined in TABLE 5.
[290] Session 9 and 10: Same as sessions 5 and 6 above, as outlined in TABLE
5, wherein the patient has the opportunity to reflect upon the MDMA session and to explore the material that emerged, and to reflect on new insights or perspectives gained/how this may impact on the issues unpinning their addictive behaviors and/or other areas of difficulty, and discuss any intentions for the next MDMA session. In addition, patients may be invited to consider any meaningful goals and/or values that they would like to focus on and move toward beyond the treatment phase. In session 10, the final session in this exemplary embodiment, the patient may complete outcome measures after the therapy session has ended. For example, this may include the Trauma History Questionnaire (THQ) (Green 1996) with the therapist. This questionnaire will give the therapy team information about the patient' s past history of trauma. The visit including the therapy session will take approximately 1.5 hours. In this embodiment, after a patient finishes the tenth visit, treatment is either considered complete, or the patient progresses to the follow-up phase, further described below.

MDMA-Assisted Psychotherapy Course¨Three MDMA Sessions
[291] In another exemplary embodiment of a MDMA-assisted psychotherapy treatment regime (also, an "MDMA-assisted psychotherapy course" or an "MDMA-assisted psychotherapy regimen"), a patient will receive 16 total therapy sessions during the treatment phase: thirteen 60-minute psychotherapy sessions, and three 6-8 hour MDMA-assisted psychotherapy sessions. Sessions will take place approximately 1-2 weeks apart and start as soon as possible after baseline (generally, within approximately one to three weeks).
!lr.A.OKU4.tiit*O.00t4.03.Tbit00-04WOOREMON184$0.0t0Nyoototopyivootkm ggiggq Week Session Activity 1 1 Non-Drug Prep Session (60-minute therapy session) 2 2 Non-Drug Prep Session (60-minute therapy session) 3 MDMA-Assisted Therapy Session 1 (-6-8 hours) 4 Next-Day Non-Drug Integration / Follow-up Session (60 mins) 4 5 Non-Drug Integration Session (60-minute therapy session) 6 Non-Drug Integration Session (60-minute therapy session) 7 MDMA-Assisted Therapy Session 2 (-6-8 hours) 8 Next-Day Non-Drug Integration / Follow-up Session (60 mins) 7 9 Non-Drug Integration Session (60-minute therapy session) 8 10 Non-Drug Integration Session (60-minute therapy session) 9 11 Non-Drug Integration Session (60-minute therapy session) 12 Non-Drug Integration Session (60-minute therapy session) 13 MDMA-Assisted Therapy Session 3 (-6-8 hours) 14 Next-Day Non-Drug Integration / Follow-up Session (60 mins) 12 15 Non-Drug Integration Session (60-minute therapy session) 13 16 Non-Drug Integration & Final Evaluation Session (-90 mins)
[292] Phases one through four preceding this 13-week MDMA-assisted psychotherapy course generally are the same as those outlined in the 8-week course of TABLE 5. Also as above, the MDMA-assisted psychotherapy sessions may be run by a therapy team, wherein the team comprises a male and a female pair, or in alternative embodiments, two males, two females, or one or two individuals not identifying with either male or female. In addition, the protocols for each therapeutic session are as generally described above, with modifications such as those in response to, or accommodation of, the overall longer course of treatment and the additional MDMA session, as will be appreciated by those of ordinary skill from the teachings herein and the general knowledge in the art.
[293] Sessions 1-9. Generally, as described above for the eight-week course of
[294] Sessions 10-12: Generally, as sessions 5 and 6 of the eight-week course of TABLE 5.
[295] Sessions 13-14: Generally, as sessions 3 and 4, and 7 and 8 of the eight-week course outlined in TABLE 5.
[296] Sessions 15 and 16: Generally, the same as sessions 9 and 10 in the eight-week course outlined in TABLE 5. Patients may continue to consider any meaningful goals and/or values that they would like to focus on and move toward beyond the treatment phase.
In session 16, the final session in this exemplary embodiment, the patient may complete outcome measures after the therapy session has ended, such as the Trauma History Questionnaire (THQ). The visit including the therapy session will take approximately 1.5 hours. In this embodiment, after patients finish the sixteenth visit, treatment is either considered complete, or the patient progresses to the follow-up phase below. In alternative embodiments, one or more of sessions 10-12, for example, are eliminated or made optional, as may be one or more other sessions, or one or more sessions in the eight week treatment protocol above (e.g., session 6 or 9).
Follow-Up and Data Collection
[297] "Follow-up" phase refers to the period following the last session of the MDMA-assisted psychotherapy course (e.g., session 10 or session 16 above) or, if a patient discontinued treatment, from two weeks after the final psychotherapy session to the final follow-up visit. In an exemplary embodiment of the treatment protocol, outcomes will be assessed at 3 months, 6 months, and 9 months after the end of baseline or the initial detoxification.
In such embodiments, the final follow-up visit is thus at 9 months post baseline or detoxification completion. In some embodiments, a final follow-up visit may be 12 months or longer post baseline or detoxification. In addition, outcomes may be assessed at other incremental timeframes, the frequency and amount of which depend on the specific needs of each participant, as would be apparent to those of skill.
[298] Means of performing follow-up visits and gathering follow-up data (e.g., to determine therapeutic efficacy and measure therapeutic effect) include in-person visits, telemedicine and other virtual visits, phone calls, automated inquiries and check-ins (e.g., email questionnaires, mobile apps, etc.), and the like. Accordingly, it will be appreciated that a "visit" need not be an in-person or face-to-face visit with a member of the therapy team, or another clinician.
[299] Audit data from clinical notes indicates that formal assessment data for patients seeking treatment for AUD (including for those who undergo community detoxification) is typically sparse and difficult to collect Accordingly, in preferred embodiments herein, means are provided to not only collect high quality data about patient outcomes, but to do so in a way that increases compliance and successful data collection. Such means will ease the burdens of collecting follow-up data (to patients, caregivers, and others), minimize attrition rates (previously shown to be in the range of 10-35% by Hallgren KA, Witkiewitz 2013), and avoid the pitfalls of using statistical analysis to predict outcomes for those lost to follow-up by collecting good quality long term outcome data in these studies from the outset, e.g., by advantageously enhancing contact with patients or by using optimized means.
[300] For example, when in person visits are used, patients will be invited to attend visits (estimated duration, 1.5 hours) at 3, 6 and 9 months post baseline or detox.
All patients will receive a reminder before the visit. At such follow-up visits, patients will be asked to complete a urine test for recreational drugs, breathalyzer test for alcohol and complete questionnaires, or other appropriate follow-up assessment. Such visits may also be conducted at other incremental timeframes, the frequency and amount of which depend on the specific needs of each participant, as would be apparent to those of skill.
[301] Follow-up telephone calls (estimated duration: 5-10 mins) also can be utilized to gather follow-up data to determine therapeutic efficacy and measure therapeutic effect. Systematic efforts will be made to contact patients to collect follow-up data; however, because some patients may be difficult to contact, patients can be asked (as an eligibility criterion) to identify a significant other who can be contacted in the event that the therapy team is unable to contact the patient to collect outcome data. When a patient is unreachable, and if the patient has consented, a significant other also may be asked to give information about outcome (drinking behavior etc.).
EXAMPLE 4: Open Label Within-Subject Safety and Tolerability Feasibility Study of MDMA-Assisted Psychotherapy in Patients with "Harmful Use" AUD
[302] In this Example is described a study to investigate the use of MDMA to treat AUD in patients without physical dependence on alcohol ("harmful use"). Patients will not require a community alcohol detoxification or similar course before their MDMA-assisted psychotherapy course. In the study of this Example, patients receive two sessions with MDMA
(e.g., 187.5 mg each session, as two separate doses totalling 125 mg and then one of 62.5 mg).
However, it should be noted, the dosage amount is purely exemplary in nature, and may be modified according to the specific outcome desired, and would be within the knowledge of one of skill. Psychological support will be provided before, during, and after each session. Safety and tolerability will be assessed alongside psychological and physiological outcome measures.
Alcohol use behavior, mental well-being, and functioning data will be collected for nine months after baseline.
[303] The study of this Example will demonstrate that MDMA treatment is well-tolerated by all participants. No unexpected adverse events will be observed. Psychosocial functioning will improve across the cohort. Regarding alcohol use, at nine months post baseline the average units of alcohol consumption by participants will compare favorably to other observational studies with similar populations of people with AUD. Such results, in view of the further description and disclosures provided herein, additionally demonstrate the therapeutic potential of MDMA-assisted psychotherapy for a population of people with non-physically dependent ("harmful use") AUD (i.e., not requiring or undergoing detox).
Methods
[304] More specifically, the study of this Example is an open label within-subject safety and tolerability feasibility study, in patients with AUD. All patients will receive MDMA-assisted therapy. The main outcome measures will be the number of patients completing the eight week psychotherapy course, the number accepting the second booster dose of MDMA on drug assisted days and adverse events. Secondary outcome measures will include changes in drinking behavior (measured by "units per week consumed," wherein a standard drink contains about 14 g of pure alcohol (ethanol), at 3 months, 6 months, and 9 months since baseline), measures of mental well-being, psycho-social functioning, quality of life and concomitant drug use.
[305] Patients with a diagnosis of alcohol use disorder, who do not have physical dependence on alcohol, will be recruited. These patients may be recruited from both NHS
and partner organisations, from primary care, by word of mouth or by answering adverts placed in appropriate locations. The sample size will be based on a power calculation of required group size to adequately test the hypothesis.
[306] After completing a group therapy preparation course, patients will receive an eight week course of abstinence-based therapy comprising 10 psychotherapy sessions (such as described in further detail above) On two of these sessions (session 3 and 7) patients will be dosed with open-label MDMA during a 6-8 hour assisted therapy session. The eight week therapeutic course will start as soon as possible after a patient has completed the preparation course (with a delay up to two weeks).
[307] In an alternative aspect, reflecting an alternative embodiment such as described above, patients will receive 16 therapy sessions, comprising 13 60-minute non-drug psychotherapy sessions and three, 6-8 hour MDMA-assisted therapy sessions. MDMA-assisted sessions will take place between three and five weeks apart (the interval between drug sessions 1 and 2 is three weeks, and the interval between drug sessions 2 and 3 is five weeks).
This 13 week therapeutic course also will start as soon as possible after a patient has completed the four week group preparation course (with a delay up to two weeks).
[308] Good Manufacturing Practice (GMP) MDMA (i.e., substantially pure racemic MDMA
HC1) will be obtained from Sterling Pharmaceuticals (Newcastle) and formulated into the investigational medicinal product (62.5 mg MDMA in gelatin capsules) by Guy's and St Thomas' Hospital's Pharmacy Manufacturing Unit (London, UK). At the time of this filing, MDMA remains a scheduled drug substance; however, the clinic holds a schedule one license for Home Office approved storage, prescribing, and dispensing of MDMA. It will be within the knowledge of one of skill as to how to obtain such a license, if it is still required.
[309] On each dosing session, patients will receive an initial oral dose of 125 mg MDMA
(i.e., two capsules), followed two-hours later by an optional booster dose of 62.5 mg (i.e., one capsule). In some embodiments, the booster dose will serve to prolong the experience, allowing for greater time for psychotherapy under the influence of MDMA. Thus, in some embodiments, a patient may only receive a single initial dose of MDMA, while in some embodiments, a patient may receive two doses of MDMA, the first being an initial dose, and the second being a booster dose.
[310] Other sessions (i.e., sessions 1, 2, 4, 5, 6, and 8-10) will comprise one hour psychotherapy sessions, and employ aspects of Motivational Interviewing and "third wave"
cognitive-behavioral approaches, for example, modified cognitive-behavioral approaches, such as Acceptance and Commitment Therapy. Patients will remain in the study for a duration of approximately 10 months.
Trial Procedures
[311] AUD will be identified using the ICD-10 SCID questionnaire. Screening will comprise of written informed consent, an evaluation of the patient's physical and mental health background, a psychiatric interview (MINI), assessments of depression and anxiety severity will be assessed using the PHQ-9 and GAD-7 questionnaires. Severity of AUD
will be established using the SADQ and SIP questionnaires. Patients will receive a thorough physical health check comprising an electrocardiogram (ECG), routine blood tests, blood pressure, heart rate, and physical examination. Following screening, eligible patients will first receive a four-week course of supportive group therapy (motivational group meetings) in order to gradually reduce their intake of alcohol. In order to confirm the patient has no symptoms of physical alcohol withdrawal, the Clinical Institute Withdrawal Assessment for Alcohol ¨
Revised Version (CIWA-Ar) (Saitz et al 1994) will be carried out, as well as an alcohol breath test. Dip-stick urinalysis for recreational drugs can be completed at the research team's discretion. If female, dip-stick urinalysis for pregnancy will be completed Patients will also be asked to complete post-preparation questionnaires.
[312] Subsequently, eligible patients enter the eight week (or 13 week) course of psychotherapy. This will entail weekly 60-minute outpatient non-drug psychotherapy sessions, delivered by two clinicians trained in delivering MDMA-assisted psychotherapy.
[313] Dosing with MDMA will occur twice during the eight week course, on weeks 3 and 6.
Physiological changes, observer and subject ratings of distress (SUDS) and the intensity of the acute psychoactive effects of MDMA will be measured throughout the MDMA-assisted session. Acute anxiety will be managed primarily psychologically, but sedative medication (oral lorazepam) will be available. Patients will optionally remain overnight in the treatment center after each MDMA-assisted session, overseen by medically-trained "night sitters," on hand to support patients as required but instructed to avoid delivering any psychotherapeutic interventions. Patients will be seen the morning after each MDMA-assisted session for an integration psychotherapy session, and then telephoned daily for six days to assess changes to mood, suicidal risk factors (using C-SSRS) and quality of sleep (using the Leeds Sleep questionnaire). Following the end of the eight week (or alternatively, 13 week) therapeutic course participants will carry out additional follow-up questionnaires. They will then be seen again at 3, 6 and 9 months (since baseline) for longer-term follow-up data collection.
Data Analysis
[314] All data will be recorded on paper case report forms (CRF s) and then digitized into MS
Excel spreadsheets. Analysis and graphing will be performed using GraphPad (Prism) or MS
Excel. When calculating Timeline Follow Back results, alcohol consumption levels at last observation will be used in the case of drop-outs or when participants undergo the group therapy preparation course more than once (Hamer and Simpson, 2009).
Results
[315] Severity of AUD criteria at screening and baseline: as per the inclusion criteria, all eligible patients will score above the diagnostic threshold on the DSM-5 SCID
questionnaire for AUD. AUD severity will also be measured using the SIP questionnaire and the SADQ
questionnaire, and no patients found to be physically dependent on alcohol will be included.
[316] Physiological and tolerability effects during MDMA sessions. Drug-assisted psychotherapy sessions with MDMA will be administered during the trial to the patients.
Temperature, BP, and FIR are measured at t=0, before taking MDMA, then half-hourly up to t=2 hours, then hourly thereafter for a minimum of six hours from the time of dosing.
[317] All of these physiological parameters are expected to remain within normal limits for all these sessions. A mild, transient rise in BP, temperature and RR over the course of the MD1VIA session may be seen. No patients will experience sustained abnormal physiological disturbance, symptomatic experiences of raised blood pressure, heart rate or temperature or any other adverse events during MDMA sessions. No medical interventions will be required in respect of these or any other physiological events during MDMA sessions.
[318] Subjective Units of Distress (SUDS) and Participant report of "Drug Effects" (as the term is used in, e.g., Sessa et al. 2021, and understood in the art) will also be measured, hourly, throughout the MDMA sessions. Most subjects are predicted to report mildly raised SUDS
scores at the beginning of the sessions before taking MDMA
_________________________ consistent with expected anxiety ahead of dosing ____________________________________________________________________ which subsequently is predicted to be reduced during the course of the session as the positive effects of MDMA emerge. Participants will give their own subjective score (0-10) of whether they felt Drug Effects, and the therapists also will record their own objective score of how "altered" the participant appeared. There will be no significant difference between Observers' and Participants' Drug Effects scores. Drug Effects will rise expectedly over the first two hours, with a further increase after the booster dose is given at t=2 hours, and a subsequent plateau then decline over the following six hours.
By the end of the MDMA session day all drug effects will have returned to baseline.
[319] Changes in drinking behavior: Data will be collected in respect of Units of Alcohol Consumed per Week (wherein a standard drink contains about 14 g of pure alcohol, i.e., ethanol) in the month before participants' baseline, immediately after baseline, throughout the eight week MDMA therapy course and for 3 months, 6 months, 9 months, one year, and over one year after baseline (including over 15 months, over 18 months, and over two years), and patients are expected to reduce their drinking behavior in significant amounts, and in amounts that significantly or markedly greater than any known or prior art treatment.
[320] Comparisons will be made for the prevalence of average number of units consumed per week between each respective time point of 3, 6, and 9 months following the final therapy session. Appropriate statistical analyses will be executed on the resulting figures via Pearsons correlation coefficients to identify the associated change between each point of TLFB data collection and baseline. The differences will then be assessed via Fisher r-z transformation (Lowry, 2001-2003) to test the hypothesis that MDMA-assisted psychotherapy compared to placebo-assisted psychotherapy is effective in this population of participants with AUD.
[321] Seven-day follow-up after MDMA sessions: Considerable medical and popular press literature reports anecdotal observation of ecstasy users experiencing an acute "come down"
effect and a drop in mood in the days after using the drug recreationally. In order to measure this prospectively with clinical MDMA, participants' mood states are measured by daily Profile Of Mood States (POMS) measurements for seven days after each MDMA session.
Positive scores represent depressed affect, zero represents no change in mood or affect, and results below zero represent a positively felt mood. Average scores across both MDMA
sessions for all participants will reveal no evidence of any mood disturbance during the week after each session of taking clinical MDMA. Participants instead will sustain a positive mood for seven days, contrasting with anecdotal reports from recreational ecstasy users.
[322] Other mental health measures and quality of life measures: Brief assessments of mood and anxiety will be made at screening, baseline, after the eight week MDMA therapy course and at 3, 6, and 9-month follow-ups, using the PHQ-9 and GAD-7 rating scales respectively. Scores are expected to demonstrate a reduction in both anxiety and depression after screening and baseline timepoints, followed by a transient rise in anxiety and depression scores 3 months after baseline and a further reduction at 6 months and a moderate rise again at 9 months post baseline. A variety of further data will be collected, including changes to the quality of sleep, quality of life measures and changes to compassion and empathy scales.
[323] Suicidality: Participants will undergo the C-SSRS rating scale at screening, baseline, throughout the eight week therapy course, in the week after each MDMA session and at 3, 6 and 9 month follow-up visits. No participants will report current suicidal ideation, intent or plans or self-harm behavior during the course of the study.
[324] Adverse Events: The acute effects of MDMA-assisted psychotherapy will be well-tolerated by participants. No unexpected adverse events will occur. No participants will report any desire to use illicit ecstasy / illicit MDMA following receiving clinical MDMA as part of this trial. No psychotic symptoms will be observed in any patients.
Analysis
[325] Through the Example, MDMA-assisted psychotherapy is demonstrated to be useful in treating AUD, and in AUD in patients who are not physically dependent on alcohol.
EXAMPLE 5: Randomized Double-Blind Between-Subject Controlled Study ofMDMA-Assisted Psychotherapy in Patients with "Harmful Use" AUD
[326] In this Example is described a randomized, double-blind controlled study of efficacy of MDMA-Assisted Psychotherapy for the treatment of patients with non-physically dependent AUD ("Harmful Use"). This is a randomized between-subject controlled study, in patients with non-physically dependent AUD. Half of the patients will receive MDMA-Assisted psychotherapy; the other half will receive placebo-assisted psychotherapy.
[327] In this study it will be shown that MDMA-assisted psychotherapy is an effective treatment for patients with AUD, who do not have physical dependence. In other words, the study of this Example will show that MDMA-assisted psychotherapy is superior to placebo dose MDMA-assisted psychotherapy and can reduce use of alcohol in patients without physical alcohol dependence and improve quality of life in patients with ADD.
[328] Outcomes regarding abstinence from alcohol, concomitant drug use (the number and frequency of both prescribed medication use and recreational drug use, including any craving related to MDMA and subsequent use of illicit ecstasy), quality of life, and psychosocial functioning will be evaluated. The main outcome measure will be efficacy, which will be measured in weekly units of alcohol consumed, (wherein a standard drink contains about 14 g of pure alcohol, i.e., ethanol) at 3, 6 and 9 months post baseline.
[329] Here, as also in Examples 4 and 8, the study will adhere to the principles outlined in the Medicines for Human Use (Clinical Trials) Regulations 2004 (SI 2004/1031), amended regulations (SI 2006/1928) and the International Conference on Harmonisation Good Clinical Practice (ICH GCP) guidelines, the Data Protection Act, and such other regulatory requirements as appropriate and as would be understood to those of ordinary skill in the art.
[330] A population of adult patients with a primary diagnosis of AUD, aged between 18 and 65, who do not have physical dependence (and therefore do not require medical detoxification) and who meet the inclusion and exclusion criteria, will be recruited (in a number sufficient to generate statistically significant results). Patients will remain in the study for a duration of approximately 14 months.
[331] As depicted in the table below, and as also further described in the other Examples herein, patients will first receive a four-week course of supportive group therapy (motivational group meetings) in order to gradually reduce their intake of alcohol. Once abstinent, they will enter a 13-week course of MDMA-assisted therapy, comprising 15 psychotherapy sessions (or, in an alternate aspect, an eight-week course as in the embodiment above).
Patients are randomized to either subtherapeutic / active control (placebo) or therapeutic dose. On three psychotherapy sessions (sessions 3, 7 and 13) patients will be given either active control (20 mg + 10 mg subtherapeutic) dose of MDMA or active therapeutic (125 mg + 62.5 mg) dose MDMA during the 6-8 hour assisted therapy sessions. Patients may optionally stay overnight, or go home after their MDMA session. Other sessions (1, 2, 4-6, 8-12, 14 and 15) will comprise non-drug-assisted 1-hour psychotherapy sessions.
[332] The sessions in general will be as described above, and reference is thus made to the description in Examples 3 and 4. From Session 15 or if discontinued treatment, from 2 weeks post the final psychotherapy session to the final follow-up visit (9 months post baseline).
Patients will be followed up at 3, 6, and 9 months from the date of baseline.
[333] A further understanding of the study procedure, including the timing and schedule of the assessments, may be obtained by reference to FIG. 17.
[334] Results are obtained as above, and by this Example, MDMA-assisted psychotherapy is further demonstrated to be useful in treating AUD, and in particular AUD in patients who are not physically dependent on alcohol, and to have such other benefits as claimed.
[335] Accordingly, and in view of all of the Examples herein, in some embodiments, the pharmaceutical compositions of the invention (and their use in the methods of the invention) are used to improve the symptoms of a mental health disorder, wherein the mental health disorder is a substance abuse disorder. In some embodiments, that substance use disorder is AUD, and specifically non-physically dependent "harmful use" AUD. The symptoms of the mental health disorder to be treated (e.g., the symptoms of non-physically dependent AUD) shall be able to be determined by one of skill, by reference to the general understanding of the art regarding that disorder, for example by reference to the DSM-5).
Accordingly, methods of the invention, and the compositions of the invention when used in those methods, will be understood as improving any such symptoms.
[336] In some embodiments, the pharmaceutical compositions of the invention (and their use in the methods of the invention) will produce a therapeutic effect, and such effect may be (1) an improvement in a symptom of a comorbid psychiatric disorder; (2) an increase in quality of life; (3) an increase in psychosocial functioning; (4) a decrease in use or frequency of prescription medication; (5) a decrease in use or frequency of recreational drugs; (6) a decrease in units of alcohol consumed; (7) a reduction of cravings relating to alcohol;
(8) a prevention of relapse.
EXAMPLE 6: Exemplary Treatment Protocols¨"Dependent Use"
[337] In certain exemplary treatment methods wherein the pharmaceutical compositions of the invention are shown to be useful in methods for treating AUD, the treatment flow can be conceived in phases as follows, and is outlined in FIG. 13: screening and initial eligibility check (1301); pre-detox group therapy, e.g., a four-week course of weekly sessions (1302); an alcohol detoxification course, e.g., over two weeks (1303); baseline visit and eligibility confirmation (1304); an MDMA-assisted psychotherapy course, e.g., over eight or 13 weeks (1305); follow-up and data collection, e.g., at 3 months, 6 months, and 9 months (1306).
[338] It will be readily appreciated, however, that this exemplary treatment flow is merely illustrative of one use of the pharmaceutical compositions in a method of treating AUD, that a use need not include every illustrated aspect (e.g., the follow-up or data collection of step six), and moreover, that the steps, stages, or phases below (and their delineation as such) are simply for purposes of assisting a reader with the conceptualization of a use of the invention, whereas the full breadth and scope thereof will be known by reference to the claims.
TABLE 8: Exemplmy Treatment Flow¨"Dependent Use"
1. Screening and Eligibility Check 2. Pre-detox Group Therapy 3. Alcohol Detoxification Course 4. Baseline Visit and Eligibility Confirmation 5. MDMA-Assisted Psychotherapy Course 6. Follow-Up and Data Collection Screening and Initial Eligibility Check (1301)
[339] Patients with AUD wishing to undergo detoxification and subsequent MDMA-assisted psychotherapy are screened prior to entering a pre-detox and detoxification program. Eligible patients will attend a baseline visit after detoxification to ensure eligibility.
[340] Potential patients are invited to a screening visit where their eligibility for MDMA-assisted psychotherapy using the methods of the invention will be determined.
The screening visit will include informed consent, medical, psychiatric and substance misuse history, prescribed medication use, basic physiological observations and alcohol breath test and urine drugs screen as clinically indicated In order to monitor the risk of patients using MDMA
outside of the therapeutic context, they can be asked about their use of, or desire to use, illicit ecstasy for example using standard questions such as used by MAPS in MDMA-assisted therapy studies.
[341] During screening the therapeutic process may be discussed and explained.
The patients' understanding of the procedure, requirements, and commitments may be confirmed and any questions answered before informed consent is obtained. If video and/or audio recordings will be made during therapy sessions, patients should be asked to consent. If patients do not wish to have sessions recorded (or at any point decide to withdraw consent for the whole or part of the session) recordings should be stopped and deleted if requested. Potential patients may be contacted and partake in a short telephone screen before invitation to a full screening visit, to minimize inconvenience to "obviously" ineligible patients_
[342] If psychiatric assessment is sought, it may be performed by a psychiatrist. Assessments may include the Mini International Neuropsychiatric Interview 5 (MINI 5) (Sheehan et al.
1998) to screen for comorbid psychiatric disorders, diagnostic evaluation of the presence of AUD according to DSM-5 (assessed using SCID-5-CT (Clinical Trials Version) (First et al.
2015)) and clinical risk assessment for suicidality using the Columbia Suicide Severity Rating Scale (C-SSRS) (Mundt 2013). Clinical data collection may include years of alcohol dependence, complications/consequences, and previous therapeutic inputs provided including any previous in-patient or community alcohol detox programs.
[343] Pre-detoxification baseline questionnaires also may be completed Information about alcohol consumption for the three months previous to the screening visit may be assessed using the Alcohol Time Line Follow Back (TLFB) method (Sobell 2001). This is a validated assessment using a calendar method and memory aids to collect retrospective estimates of daily drinking. Patients also may be given the same calendar to take away to help track their drinking behavior for the duration of the therapy.
Pre-Detox Motivational Group Therapy Phase (13021
[344] After screening, patients attend four weekly sessions of motivational or supportive group therapy pre-detox, e.g., run by a trained counsellor with expertise in addictions. The purpose of the group will be to provide psycho-social support to allow adequate preparation and consideration of appropriate aftercare for individuals with AUD wishing to have a detox.
Pre-detox psycho-social work benefits from consideration of a person's support network, motivation and after-care planning to improve detoxification outcomes (Kouimtsidi s, 2017).
[345] Groups will be, e.g., between five and 10 patients and may be facilitated by one or two trained staff with experience in delivering pre-detox group counselling. Each group session may be for, e.g., 60 minutes and should take place at a predetermined location. In some embodiments, pre-detox group sessions will last for about 30 minutes, about 35 minutes, about 40 minutes, about 45 minutes, about 50 minutes, about 55 minutes, about 60 minutes, about 65 minutes, about 70 minutes, about 75 minutes, about 80 minutes, about 85 minutes, about 90 minutes, about 95 minutes, about 100 minutes, about 105 minutes, about 110 minutes, about 115 minutes, about 120 minutes, and values in between. In some embodiments, pre-detox group sessions may last for greater than 120 minutes, or may take place in a residential (or -rehab") facility, In some embodiments, groups are closed and confidential. Patients may be enrolled into the groups on a rolling basis, and will leave after a minimum (e.g., of four) weeks attendance to be enrolled into a detox course when deemed appropriate by the facilitators. If a patient fails to attend at least (in the example embodiment, four weeks) of pre-detox group counselling, their detox may be delayed until deemed ready to progress.
Similarly, if a patient is not deemed sufficiently engaged in the preparation phase¨generally, as deemed by the group facilitators¨their detox may be delayed until they are considered to be adequately prepared (e.g., by attending further group sessions).
Alcohol Detoxification Phase (1303)
[346] Alcohol detoxification may be achieved either by gradual reductions in daily alcohol use (which may begin from the start of the pre-detox group phase, with support from the group facilitators) and/or through a medical detox. The medical detox procedure, outlined in FIG 3., is disclosed in further detail as follows.
[347] First, a patient will undergo a medical assessment (1401). Physical examination and baseline physiological measures, e.g., blood pressure, will be recorded.
[348] Second, and importantly, the patient will stop daily drinking (1402).
[349] Third, the patient may attend a clinic daily (1403) to be prescribed a sliding scale of benzodiazepine treatment (such as chlordiazepoxide) over the next 10-14 days.
Chlordiazepoxide or other pharmacotherapeutic treatment (e.g., another benzodiazepine) is prescribed according to good medical practice and dosing regimens known to those in the art.
[350] Fourth, the patient may be assessed by a specialist addictions nurse at each daily visit throughout the detoxification (1404). Daily measurements of blood pressure may be performed.
Daily measurements of the Clinical Institute Withdrawal Assessment for Alcohol¨Revised Version (CIWA¨Ar) (Saitz et al. 1994) (detox assessment measure) may be used to assess response to benzodiazepine administration and progress of the detox.
[351] Fifth, after 10-14 days (depending on the results of the CIWA¨Ar and starting dose of chlordiazepoxide or other pharmacotherapy used), the patient is assessed for success of the detox (1405). A medical opinion is sought to assess suitability to progress into treatment.
[352] If the detox is found to be successful (1406)(1407), the detox is complete (1408) and the patient may proceed to treatment. If the detox is found to be incomplete, and thus not successful (1406)(1409) the clinical team may decide whether further interventions are required (1410) before progressing to MDMA-assisted psychotherapy, or whether the patient must return to a prior step (1411).
Baseline Visit and Eligibility Confirmation (1304)
[353] After detoxification is completed, patients may attend a baseline visit to confirm eligibility for continuation into MDMA-assisted psychotherapy. During this visit, continued consent should be confirmed and vital signs can be reassessed A brief physical examination and ECG may be performed, and blood samples for routine laboratory tests (urea and electrolytes, full blood count, liver function tests and Gamma-CT) also may be completed. A
blood test may not be required if the patient has had one in, e.g., the last three months.
[354] In order to confirm the patient has adequately overcome the symptoms of physical alcohol withdrawal, the Clinical Institute Withdrawal Assessment for Alcohol¨Revised Version (CIWA¨Ar) (Saitz et al. 1994) may be carried out, as well as an alcohol breath test.
Dip-stick urinalysis for drugs of abuse can be completed. If female, dip-stick urinalysis for pregnancy may be completed. Patients may be asked to complete post-detoxification questionnaires. Patients may generally start MDMA-assisted psychotherapy as soon as possible after completion of detoxification (generally, within approximately 1 to 3 weeks).
[355] As above, the MDMA-assisted therapy course may be an eight-week MDMA-assisted psychotherapy course with two MDMA sessions (as in TABLE 5) or a 13-week MDMA-assisted psychotherapy course with three MDMA sessions (as in TABLE 6), or a modification of either, based on the teachings herein and the general knowledge in the art.
EXAMPLE 7: Protocol for MDMA-assisted therapy for A LID
[356] Attention should now be paid to FIG. 16, which outlines one embodiment of the protocol of this Example that is useful in treating alcohol use disorder in a patient in need thereof, and is further discussed below. In some embodiments, the protocol of this Example will be used with one or more individuals (the term used interchangeably with patients) who have harmful use AUD; in other embodiments, the protocol of this Example will be used with one or more individuals who have dependent use AUD; in other embodiments, the protocol of this Example will be used with multiple individuals, some having harmful use AUD, and some having dependent use AUD; in all such embodiments, modifications are made as will be readily appreciated by those in the art (e.g., substituting Pre-detox Group Therapy and Pre-MDMA
Course Preparatory Group Therapy Course with Alcohol Detoxification Course).
[357] A practitioner will first confirm the patient has alcohol use disorder (1601). This may be accomplished via assessing whether the individual meets at least 2 of the 11 listed criteria for AUD in the DSM-V during the same 12-month period, the criteria being whether the individual has: "(1) Had times when you [i.e., the patient] ended up drinking more, or longer, than you intended? (2) More than once wanted to cut down or stop drinking, or tried to, but couldn't? (3) Spent a lot of time drinking? Or being sick or getting over other aftereffects? (4) Wanted a drink so badly you couldn't think of anything else? (5) Found that drinking¨or being sick from drinking¨often interfered with taking care of your home or family?
Or caused job troubles? Or school problems? (6) Continued to drink even though it was causing trouble with your family or friends? (7) Given up or cut back on activities that were important or interesting to you, or gave you pleasure, in order to drink? (8) More than once gotten into situations while or after drinking that increased your chances of getting hurt (such as driving, swimming, using machinery, walking in a dangerous area, or having unsafe sex)? (9) Continued to drink even though it was making you feel depressed or anxious or adding to another health problem? Or after having had a memory blackout? (10) Had to drink much more than you once did to get the effect you want? Or found that your usual number of drinks had much less effect than before? (11) Found that when the effects of alcohol were wearing off, you had withdrawal symptoms, such as trouble sleeping, shakiness, restlessness, nausea, sweating, a racing heart, or a seizure? Or sensed things that were not there?"
[358] A screening visit is conducted (1602), wherein a determination will be made as to whether or not the individual is fit to undergo treatment. Whether or not the individual is fit to undergo treatment will be determined by obtaining objective measurements, such as using inclusion and exclusion criteria as discussed below and set forth herein, or additionally and optionally including any regarding the individual's weight, body temperature, HR, respiratory rate, blood oxygenation, BP and its variables, including, but not limited to:
SBP, SBP, MAP, and PP; CNIBP; ECG measurements, including RR interval or its variability, QT
interval or its variability, 1-[RV (including measured by devices other than an ECG);
hemodynamic response, and levels of glucose, cortisol, serotonin, dopamine, cholesterol; EEG
measures; BDNI4;
genetic markers including relating to CYP enzymes or drug metabolism; genetic variation in mGluR5 or FKBP5; and numerous more, as known to those in the art.
[359] The screening visit will also include assessing whether or not the individual meets all inclusion criteria (1603). As disclosed herein, the inclusion criteria include informed consent, a primary diagnosis of AUD, being between the ages of 18 and 65 years old, identifying an individual able to accompany the patient to at least one study visit if required and be contacted by the study team in the tevent the patient could not be contacted, being proficient in speaking and in reading English, and agreeing to comply with the requirements of the protocol. Such inclusion criteria may be assessed by evaluating an individual's medical, psychiatric, and substance misuse history; prescribed medication use, basic physiological observations and alcohol breath test, and urine drugs screen, or others as clinically indicated.
[360] In addition to meeting all inclusion criteria (1603) an individual in some embodiments will also not have not met at least one exclusion criteria, wherein the exclusion criteria include any one or more of lacking capacity, having a history of a primary psychotic disorder, bipolar affective disorder type 1, or a personality disorder; posing a serious suicide risk, having at least one abnormal clinical finding during the screening visit, being a regular user of ecstasy, currently taking medication likely to interact with MDMA, being a regular use of other drugs or having a dependence thereon (besides also alcohol), being pregnant, breastfeeding, having taken part in a study involving an investigational product in the last three months, having an immunological disease, havingunstable hypertension, havingsevere liver disease, having cardiac disease, having an active infection, and having had an infection within four weeks of MDMA administration.
[361] Where all inclusion criteria are met (1603) and no exclusion criteria are met (1604), in such embodiments, an individual may proceed as a patient and complete the four-week preparation group course (1605). As discussed below, the four-week preparation group course may employ group-therapy tactics to enable an individual to lessen their reliance on alcohol;
"reliance" may refer to using alcohol as a means to cope with negative experiences in daily life. The exemplary course described here will broadly cover four separate topics, including confidence building; resources, barriers, and goal setting; increasing rewarding activities and building support, and building the foundations for recovery.
[362] So long as the patient completes the preparation group course (i.e., actively participates and does not resume alcohol consumption), the patient then completes an 11-week therapy protocol comprising 14 psychotherapy sessions (under the supervision of a therapist pair or "therapist team"), three of which are MDMA-assisted psychotherapy sessions (lasting from between about 360 minutes to about 480 minutes), wherein the patient is administered an initial dose of between about 100 mg to about 150 mg of MDMA, and optionally a booster dose of about half of the initial dose (thus, if the initial dose was about 100 mg, the booster dose is about 50 mg); and 11 of which are about 60 minute non-drug psychotherapy sessions (1606).
In some embodiments, the booster dose is administered to prolong the MDMA
experience. In some embodiments, the booster dose is administered to increase the time with which the patient and the therapist pair have to interact while the patient is under the effects of MDMA.
[363] Once all 14 psychotherapy sessions have been completed, the patient is further assessed at a visit three months, six months, and nine months after the conclusion of the therapy protocol (1607), or otherwise following treatment as defined below. Patients may also be assessed at other times following treatment, such as at one month, at one year, at greater than one year, at times in between, and at points periodically, semi-continuously, or continuously (e.g., by mobile apps, fitness trackers, regular email correspondence, and the like). In some embodiments, the amount of alcohol consumed per patient at the three, six, and nine month visits will be less than the alcohol the patient consumed prior to beginning the protocol of this Example. In some such embodiments, the amount of alcohol consumed per patient is reduced (i.e., measured at any one or more of the three, six, and/or nine month visits) will be less than the alcohol the patient consumed prior to the start of the preparation group or prior to the start of detox, or otherwise prior to undergoing the method of the invention (e.g., when measured at or before intake). In some preferred embodiments, the amount of alcohol consumed per patient at the three, six, and/or nine month visits will be 50% or less of the alcohol the patient consumed prior to the start of the preparation group or prior to the start of detox, or otherwise prior to undergoing the method of the invention. In some preferred embodiments, the amount of alcohol consumed per patient at the three, six, and nine month visits will be a reduction of at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, up to and including a reduction of 100%, i.e., a patient is no longer consuming alcohol. Other therapeutic effects or outcomes which a patient may experience will be as discussed below.
[364] Regarding the preparation group course in more depth, if eligible at screening (which includes assessing whether the individual meets all inclusion criteria, as discussed herein, and does not meet at least one exclusion criteria, as discussed herein; as well as ensuring the individual is able to undergo treatment, which is assessed by collecting objective measurements including but not limited to weight, body temperature, heart rate, respiratory rate, blood oxygenation, BP and its variables, including SBP, SBP, MAP, and PP; CNIBP; ECG

measurements, including RR interval or its variability, QT interval or its variability, HRV
(including measured by devices other than an ECG); hemodynamic response, and levels of glucose, cortisol, serotonin, dopamine, cholesterol; EEG measures; BDNF;
genetic markers including relating to CYP enzymes or drug metabolism; genetic variation in mGluR5 or FKBP5; and numerous more, as will be readily appreciated by those of skill);
the participant will be enrolled in a four-week preparation group course to support them with making changes to their problematic alcohol use. Each session lasts for 60 minutes and draws on motivational interviewing approaches, as detailed herein. It is anticipated that participants will further benefit from the peer support elements offered by the group format. As mentioned, this will be a rolling group, meaning participants can join at any week and attend for at least 4 weeks.
[365] The content of the preparation group course will broadly cover four topics:
1. Building confidence a. Discussing problems drinking has caused, with family, job, social life, health, etc.
b. Why change is necessary (completing a cost benefit analysis worksheet) c. Building confidence, discussing when something went well for the patient d. Completing a drinking diary, where drinking is monitored daily for one week e. Completing "homework" tasks, such as the drinking diary, and a worksheet detailing strengths and weaknesses of the patient, completed by the patient 2. Resources, barriers, and goal setting a. Discussing goal setting, specifically having participants elicit goal setting ideas.
Exemplary goals may include setting non-drinking days, changing to lower alcohol drinks, increasing the time between drinks, drinking water in between drinks, etc.
b. Reviewing potential barriers and resources that may aid in the reduction of alcohol consumption, such as discussing the patient's strengths and qualities, and identifying social and community support outlets c. Completing homework tasks, such as setting a goal related to alcohol consumption, completing a daily diary, and completing a worksheet that details social and community support outlets 3. Increasing rewarding activities and building support a. Identifying challenges in achieving goals, and discussing how those challenges were/are managed b. Reviewing the social and community support worksheet, and working as a group to build upon the resources listed within c. Setting rewarding goals, including planning pleasant activities, etc.
d. Completing homework tasks, which may include setting a goal related to drinking, and beginning a pleasant activities planner 4. Building the foundations for recovery a. Discussing how the patient will manage moving forward, including identifying anticipated challenges and brainstorming ways to manage those, and completing a "planning for the future" worksheet b. Identifying any changes the patient would like to make in their recovery program, and completing a worksheet identifying such changes, if so c. The patient then completes a recovery support map, and shares/discusses the map with someone who supports the patient
[366] As mentioned, so long as the patient adequately engages in the preparation course, remains eligible (i.e., satisfies the aforementioned inclusion criteria), and illustrates a motivation to make changes to alcohol use during the preparatory course, the participant will commence a 14 session course of MDMA-assisted therapy as soon as possible. The investigators will aim to start the course within one week if possible.
[367] Regarding the 14 session, 11 week therapy protocol, all sessions will be conducted by the same two therapists per participant. There will be a total of three 'drug' sessions (6-8 hour MDMA facilitated) wherein an initial dose of about 120 mg and, optionally, a booster dose of about 60 mg are administered, and 11 'non-drug' (60 minute talking therapy) sessions. Non-drug sessions may be conducted by one therapist on rare occasions if this is unavoidable. All drug sessions must be facilitated by two therapists and the MDMA capsules must be administered by a medical doctor. One therapist at a time may leave the therapy room for brief periods (e.g., for comfort breaks). The participant should be accompanied for the duration of the session and for 6 hours after initial dosing.
[368] All participants starting the main therapy course will have worked on reducing their alcohol intake over the 4-week preparatory group program. Therefore, the main aims of the subsequent 14 session therapy course are to support participants to maintain/build upon these changes, prevent relapse into heavy drinking and develop skills to lead a fulfilling life without problematic drinking.
[369] Broadly, the three MDMA-assisted psychotherapy sessions will be conducted at weeks 3, 6, and 9. As mentioned, these sessions will last for approximately 6 to 8 hours (360 to 480 minutes), while the non-dosing physiotherapy sessions (which occur all 11 weeks) will last for approximately 1 hour (60 minutes). The therapy appointments prior to the participant's drug assisted sessions will give the therapist team and participant the opportunity to develop a good therapeutic alliance, which is also benefited by the participant having the opportunity to tell their story. This helps to create a collaborative understanding of problem development and maintenance, alongside identification of resources and protective factors.
These early sessions give the participant an opportunity to ask any questions about the drug experience, to set an intention for what they would like to get from this experience, and for therapists to describe what they can expect in terms of practicalities and support on the day. The drug-assisted session content is led by the participant, with the therapists in a supporting role and occasionally making gentle suggestions as appropriate. Subsequent integration sessions (i.e., non-drug psychotherapy sessions) arc an opportunity to reflect on the MDMA experience and to consider any insights and new perspectives that may be emerging and how these may be meaningful to the participant's day to day life
[370] The 14-week, 11-session therapy protocol (also an "MDMA-assisted psychotherapy course- or "MDMA-assisted psychotherapy regimen") is disclosed in the table below:
TABLE 9: Description of Session Content for Exemplar), MDMA for AUD Program Week Visit Session Description of Session Content No. No. No.
Introduction, preparation, start of participant's story, therapy 3 1 goals and introduction to intention setting.
60 Introduction to the therapy program (relapse prevention embedded in a mins framework of enhancing psychological flexibility, using MDMA-assisted therapy to support access to and processing of the factors that have contributed to the development and maintenance of problematic alcohol u s e) -Discuss the role of MDMA as an adjunct to psychotherapy.
Opportunity to discuss any questions or concerns that arise.
-Participant's personal story¨contributing and maintaining factors relevant to alcohol use, strengths and resources (strengths and resources to be discussed in more detail in subsequent sessions).

-Discuss participant's overall goals for therapy and introduce idea of intention setting for their first MDMA assisted session Continue preparation, intention setting, teach and practice 2 4 2 relaxation technique, review of current recovery plan.
60 -Continue to discuss and develop a shared understanding of lillS participant's personal story.
-Discuss and plan intention for MDMA-assisted session.
-Practice at least one relaxation/anxiety management technique that can be used in the MDMA-assisted session. Discuss use of interpersonal touch for support during sessions and whether participant is comfortable with this.
-Review of recovery support map developed during preparation group.
Drug-assisted session 1. Therapist's follow participant's lead. No 3 5 3 new material introduced by therapists.
Journaling encouraged after the session.

hours -120 mg MDMA + 60 mg MDMA booster 1.5 to 2 hours later.
-The participant will always be accompanied by at least one therapist for the duration of the drug-assisted session. Each therapist may leave the room for short periods for comfort breaks but the majority of the session will be conducted with both therapists present.
-After taking the initial dose (overseen by a medical doctor), the participant will be invited to relax on the bed and listen to music through headphones and with eyeshades on. Therapists will typically make occasional check ins with the participant, usually at the same time that vital signs are taken.
-Between 90 minutes and two hours after receiving the initial dose, the participant will be given the option to receive a further supplemental dose of half the original dose (60mg of MDMA). The purpose of this is to prolong the psychological and direct pharmacological effects of the drug to allow for further processing to occur. This dosing and pattern of dosing regime is standard for MDMA-assisted psychotherapy, and has been employed in previous MDMA-assisted psychotherapy pilot studies (Mitheofer 2011, Oehen 2012).
-Participants are encouraged to have an internal experience to a large extent during this session and spend reasonable amounts of time 'inside' with music playing through headphones and eyeshades on. If the participant wishes to talk during this session, this is led by them, with the therapists following their lead and responding with open, interested questions. At certain times, the therapists may encourage the participant to return to having an internal experience, to see what comes up for processing. The participant is not encouraged to get up and move around during the session, but rather to remain sitting or lying throughout. However, if a patient is keen to get up (e.g., they need to go to the bathroom), this can be facilitated if the study team judge this as safe to do so.
-Towards the end of the session, the participant may wish to talk more as they become more orientated towards their external environment.
Once the participant feels ready, the therapy session can be drawn to a close and the participant can have some quiet time to reflect, rest and take light refreshments. Their significant other will be contacted to escort them home no less than 6 hours after the initial MDMA dose.
-Journaling to facilitate on-going processing is given as a home task.
Integration of MDMA experience. Introduction to mindfulness 6 4 and mindfulness practice.
60 -The participant may choose to provide a narrative of this experience mins and the therapists will ask open-ended questions to support the participant to draw personal meanings and insights and to consider how this may impact on their behaviour going forward.
-Mindfulness exercise to finish (mindful body scan).
Ongoing integration from medication-assisted session, High Risk 4 7 5 Situations for relapse and responses.
60 -Continue discussion re processing of material from the MDMA
mins session.
-Discuss the workability of alcohol as a control strategy.
-Discuss potential High Risk Situations (HRS) for relapse and workable responses (including individual strengths and network and community resources).
Understanding how our thoughts hook us in.

-Identify fusion with thoughts (getting 'hooked'): rules, reasons, 60 judgements, past, future, self.
mins -Introduce defusion techniques (getting 'unhooked') and practice defusion exercises.
-Noticing hooks and practising defusion from thoughts set as home task Drug-assisted session 2. Therapist's follow participant's lead. No 6 9 7 new material introduced by therapists.
Journaling encouraged after the session.

hours -120 mg MDMA + 60 mg MDMA booster 1.5 to 2 hours later.
-refer to session 3 for details 8 Integration of MDMA experience. Mindfulness practice.
60 -The participant may choose to provide a narrative of this experience mins and the therapists will ask open-ended questions to support the participant to draw personal meanings and insights and to consider how this may impact on their behaviour going forward.
-Mindfulness exercise to finish (mindful breathing).
Opening up by letting go of the struggle -Identify problematic experiential avoidance of internal experiences.
mins -Introduce and practice acceptance techniques.
-Discuss intention for second MDMA-assisted session.
-Noticing avoidance and practicing acceptance of emotions as home task.
Values and committed action -Introduce values (as different to goals) and elicit values from the 60 participant.
mins -What's important in their life? What they like to do more of/less of?
What strengths do they already have to achieve this and which ones would they like to develop?
-Consider immediate, short, medium and long term goal setting in accordance with values.
-Identify barriers (FEAR; Fusion, Excessive goals, Avoidance of discomfort, Remoteness from values) and potential responses to barriers (DARE; Defusion, Acceptance of discomfort, Realistic goals, Embracing values).
-Complete a short-term SMART goal for home task.
Drug-assisted session 3. Therapist's follow participant's lead. No new 9 13 11 material introduced by therapists. Journaling encouraged after the session.

hours -120 mg MDMA + 60 mg MDMA booster 1.5 to 2 hours later.
-refer to session 3 for details.
Integration of MDMA experience. Mindfulness practice.

-The participant may choose to provide a narrative of this experience 60 and the therapists will ask open-ended questions to support the mins participant to draw personal meanings and insights and to consider how this may impact on their behaviour going forward.
-Mindfulness exercise to finish (compassionate body scan).
Stepping out of auto-pilot, developing self-compassion and 15 13 responding to lapses.
60 -Noticing urges to drink alcohol by stepping out of autopilot. Relate to mins Seemingly Irrelevant Decisions (SIDs).
-Discuss everyday mindfulness practice to increase purposeful awareness.
-Introduce and practice SOBER breathing space.
-Discussion of inner critic and self-compassionate response.
-Build on self-compassion work to start lapse response plan.
-Everyday mindfulness and completion of lapse response plan as home task.
Review of the therapy course and ongoing recovery planning.

-Plans for on-going recovery and integration work within the 60 participant's community.
mins -Review of recovery plan
[371] As it relates to the therapy sessions themselves, as mentioned, there are a total of 14 sessions spanning 11 weeks, wherein 3 of the 14 sessions are MDMA-dosing sessions wherein a patient is administered an initial dose, and optionally a booster dose, the initial dose comprising between about 100 mg to about 150 mg, and the booster dose comprising about half of the initial dose, such that¨if the initial dose is about 100 mg of MDMA, the booster dose would be about 50 mg of MDMA. In other words, the proportion of the initial dose as compared to the booster dose is 2:1, wherein the initial dose is twice as much as the booster dose. As stated herein, the booster dose is an "optional" dose, wherein the booster dose is not required and, in some embodiments of the invention, the therapy protocol includes three MDMA-assisted psychotherapy sessions, wherein the optional booster dose is not administered to the patient. Likewise, in some embodiments, the optional booster dose is administered during one of the MDMA-assisted psychotherapy sessions; in some embodiments, the optional booster dose is administered during two of the MDMA-assisted psychotherapy sessions;
and, finally, in some embodiments, the optional booster dose is administered during all of the MDMA-assisted psychotherapy sessions.
[372] In some embodiments, the optional booster dose is administered to prolong the subjective experience felt by the patient when administered an effective dose of MDMA. In some embodiments, the optional booster dose is administered to allow for additional time for the patient and therapist team to discuss and work through any internalized emotions or feelings that surface for the patient.
[373] In some embodiments, each of the 3 MDMA-assisted psychotherapy sessions will last for between about 360 minutes to about 480 minutes. In some embodiments, the non-dosing therapy sessions will last for about 60 minutes.
[374] Further information about individual sessions, in an exemplary embodiment, follows.
TABLE 10: Sumniary of Exemplaty MDMA for A UD Program Week 1 = Introduction: Overall MDMA for AUD Program and goals Session 1 = Start to develop a shared understanding of your story in the context of alcohol use = Start preparation for your first medication-assisted session Week 2 = Continue to develop a shared understanding of your story in the context of alcohol use Session 2 = Continue preparation for your first medication-assisted session = Review of recovery plan developed in preparation group Week 3 = Ready for your first experiential session Session 3 = Journaling Session 4 = Introducing mindfulness and practice Week 4 = Opportunity to continue discussion regarding processing of material that emerged during experiential session.
Session 5 = High Risk Situations (HRS) and coping responses Week 5 = Unhooking from your thoughts Session 6 = Preparation for your second experiential session Week 6 = Ready for your second experiential session Journaling Session 7 = Mindfulness practice Session 8 Week 7 = Opportunity to continue discussion regarding processing of material that emerged during experiential session.
Session 9 = Opening up by letting go of the struggle Week 8 = What matters: identifying personal values Session 10 = Goal setting in accordance with values Identifying barriers and responses Week 9 = Ready for your third experiential session Session 11 Session 12 = Journaling = Mindfulness practice Week 10 = Ready for your third experiential session Session 13 = Journaling = Mindfulness practice Week 11 = Review of the therapy course: Putting it all together Session 14 Week 1, Session 1
[375] The first weekly session will serve as an introduction to the MDMA for AUD therapy protocol, and will cover the purpose of the study (i.e., whether MDMA-assisted psychotherapy is more effective for preventing relapse to harmful drinking compared to psychotherapy without MDMA), and will provide a general overview of the protocol, including that the sessions will focus on reduction strategies and promoting psychological flexibility.
[376] The participant will then be invited to ask any questions. If the participant asks about safety, the administration of MDMA in thousands of modern studies across the world will be discussed, along with the fact that the drug is considered very safe when delivered in combination with therapy and in a highly controlled setting where appropriate monitoring is in place.
[377] The therapy team then discusses with the patient how MDMA works as an adjunct to psychotherapy, which aids in developing a collaborative "formulation," which is a provisional explanation or hypothesis of how an individual comes to present with a certain difficulty or circumstance at a particular point in time (Weerasekera, 1996) As it relates to the invention, a formulation should be a collaborative process between the patient and therapist team that will be added to, and altered, as the therapeutic process unfolds. It can be considered as a "road map" that will inform the direction of the therapy.
[378] When meeting with a patient for assessment, a psychiatrist and a psychologically informed practitioner will jointly assess their suitability, and in doing so seek to understand their situation with the use of a formulation based around the 5 Ps (Macneil, et al, 2012):
(1) What is the Problem that brought you here? (2) What are the Predisposing factors? E.g., life events, family history, traumas, etc. (3) What Precipitated you seeking help now? e.g., recent triggers. (4) What Perpetuates the problem? E.g. thoughts, feelings and behaviours that keep the problem going. (5) What Protective factors are there? E.g. what positives, strengths or assets the person has got going for them. Another formulation can focus on (1) Current difficulties; (2) Past difficulties; (3) Triggers; (4) Maintenance factors (physical, social or psychological factors); and (5) Protective factors (strengths, resources, people or services that are supportive, things learned from experiences, hobbies, etc.).
[379] During the assessment session, the clinicians will listen to the patient's history through the lens of ACT (Acceptance and Commitment Therapy), noticing examples of cognitive fusion, experiential avoidance, loss of values or other examples of psychological inflexibility.
This will help identify areas on the ACT hexaflex as having potential for change for the individual. If it is determined that the patient is suitable for the treatment, the case conceptualisation will be further developed with the use of an ACT-based formulation that will guide the therapist and patient in achieving greater psychological flexibility (see below).
[380] The therapy team and patient will then work together to form a shared understanding of the formulation, which includes the identification of difficulties, and areas of strength and resource. The therapy team and patient will also work to develop therapy goals for the patient, and introduce "intention setting,- where the patient sets an intention for the drug-assisted session. As described by Mithoefer (2017), it is useful to invite a patient to have a focus for the session but to hold this lightly so that they do not grip onto the intention so tightly it becomes restrictive and potentially disruptive to the natural process that unfolds.
Rather, they have a focus in mind but are prepared to go with whatever comes up for them.
[381] Subsequent integration sessions are a valuable opportunity to build upon the insights gained during the drug experience, and to consider how these may translate into meaningful and lasting changes in accordance with what the individual feels really matters to them in their life. The therapeutic model for these integration sessions will be drawn from principles of ACT
in order to promote and enhance ongoing psychological flexibility that has been initiated by the drug-assisted therapy sessions.
[382] The therapy team will then discuss with the patient the use of ACT in the integration process. Broadly, the goal of combining drug-assisted therapy with ACT is to provide a therapeutic framework that will enable patients to alter patterns of cognition, feelings and behavior that create unworkable actions, and at the same time increasing engagement in line with the values that are important to them. The interaction between these two mechanisms of change (drug-assisted therapy and ACT) provide the environment to increase psychological flexibility and help improve the patient's life.
[383] One premise is that drug-assisted therapy requires the active engagement of patients in their own healing, assisted by a therapeutic framework. This is in contrast to the idea of psychedelic therapy as a "magic bullet- intervention, whereby the administration of the drug causes the appearance of an "inner healer" to do the necessary repair work unassisted. ACT
provides a platform by which a patient can actively engage in the process of recovery through specific practices informed by the hexaflex, which leads to increased psychological flexibility, accentuated by the effects of MDMA which allows for enhancement of neuronal plasticity. It is anticipated that the combination of drug effects and psychotherapy provide the optimal conditions for the patient to work on their recovery by enhancing the patient's own innate ability to heal themselves.
[384] ACT encourages acceptance of internal and external discomfort and an ability to tolerate and fully experience them with openness, thereby decreasing the need to use coping strategies of experiential avoidance and unworkable actions. The ultimate aim is to live a full, meaningful life, which will indirectly lead to a reduction in the mental health symptoms that they present with, as well as reducing other associated problematic behaviours.
[385] Following the initial preparation stage the therapy protocol alternates between drug-assisted therapy sessions and ACT-informed integration sessions based on the hexaflex;
specifically to: (a) be in the present moment; (b) develop a more flexible experience of the self (self-as-context) rather than be fused to a particular personal narrative (self-as-content); (c) disengage from attempts to control thoughts and emotions and instead, observe and accept them as they are (acceptance and defusion), (d) clarify values in the areas of their life they would like to prioritize; and (e) engage in committed action in line with their values.
[386] The ACT-based interventions involve a combination of techniques that enhance the effects of the drug-assisted dosing sessions in order to achieve greater psychological flexibility.
These techniques and processes involve a synthesis of several factors in line with ACT:
theoretical explanations about ACT principles, metaphors to illustrate key concepts, the use of worksheets; experiential exercises within session; revisiting the ACT specific formulation; and homework tasks between sessions. A guide with suggested exercises and the order in which these processes may be delivered can be provided. A participant will be asked to bring along their recovery support map developed during the preparation group to session 2.
Week 2, Session 2
[387] The second session will generally continue from the first, building on the topics introduced in week one. If the development of the collaborative formulation is not completed during session 1, it is done so in session two. The therapist team will also practice at least one support technique and discuss/practice the use of "interpersonal touch" with the patient during this session.
[388] A key skill of psychedelic-assisted psychotherapy work is to establish excellent patient-therapist rapport as quickly as possible. This will help the patient to feel comfortable enough in their drug-assisted session to be present and interested in whatever comes up for them, rather than trying to avoid or turn away from it. The therapist should spend a few minutes discussing supportive/grounding techniques during preparation; exploring whether the patient has any current ways of managing emotional discomfort that could be useful for the session. Make it clear to the patient that the aim of using a supportive technique is to help the patient to persevere through, rather than avoid, any challenging experiences as much as possible.
Discuss the use of supportive touch and whether the patient would feel comfortable with this.
Clarify boundaries, all touch is non-sexual and will be on the shoulder, arm or hand.
Practice in preparation how the patient may reach out for supportive touch and disengage from the same.

It is useful to have an agreement and practice beforehand in the event of challenging experiences arising that the patient would appreciate support with in order to remain in, rather than move away from, the experience.
[389] The therapist team will also discuss supportive/grounding techniques with the patient, non-limiting examples of which include diaphragmatic breathing, wherein the patient is instructed to "take a long, full breath from your abdomen, rather than from your chest In through your nose and out through your mouth. As you breathe, notice any sensations, thoughts, and images as they come and go as you gently breathe in and out from your tummy;"
as well as imagery, wherein the patient is instructed to "close or lower your eyes. Invite a calm and relaxing scene to come to mind and try to make it as vivid as possible.
Allow yourself to wander and fill in the details of the image." The therapist may also utilize the following follow-up prompts: "What can you see/smell/feel? How does it feel to you? Where do you notice it in your body? What's it like?" If the patient appears to be distressed, the practitioner may also instruct the patient to "breathe with me¨in through the nose, out through your mouth. Think of a place where you feel safe (can make suggestions if needed), imagine how safe and relaxed you feel, think of it in as much detail as you can. Describe it to me if you like.- Regardless of the method applied, the most important aspect is to ensure the patient is grounded such that their immediate environment can be used if the patient's distress appears to be overwhelming.
In extreme situations, at least one of the therapist pair may use reassuring phrases, such as "you're here with us and you're safe, we're right here with you," and may distract the patient, such that they are focused on external surroundings rather than internal feelings, including exemplary phrases such as "how does the couch feel as you lay on it?" "what can you see around the room?" "are there any noises you can hear?" Note, this technique should be used after other options such as supportive touch (e.g., hand holding), use of the breath, etc.
[390] If the non-drug sessions have taken place in a different room to the one that will be used on dosing day, ensure that the patient is able to see the room before session 3.
[391] The therapist pair will then briefly introduce to the patient what is meant by "psychological flexibility" in terms of three key concepts: "open up"
(defusion & acceptance), "be present" (contact with present moment & self as context), and "do what matters" (values & committed action). The therapist pair is not required to spend extensive time explaining these concepts in detail, but can rather introduce them to the patient and explain how they are associated with reduced suffering and enhanced quality of life (rather than symptom reduction necessarily).
[392] The therapist pair will then review the recovery plan developed in the preparation group with the patient, discussing key elements and requesting the patient discuss with them any misapprehended points. The therapist pair will then inquire about what seems to be working well, anything that may be particularly challenging, anything the patient feels could be added, and/or anything the patient would like to develop/change at this point. The therapist pair will ensure they have the opportunity to review and add to the recovery plan with the patient during the final session, when the patient will have learned and practiced skills that may be new to them, and that they may wish to continue developing as part of their ongoing recovery.
[393] At this point, patients may be introduced to the concept of maintaining an experiential therapy journal where they are encouraged to write down anything they find significant during their experiential day. For example, any impactful emotions, perspectives, images, questions, or thoughts that arose. These can be discussed in integration sessions with the therapist pair.
Week 3, Session 3
[394] For the first dosing session, the therapist pair will arrive in advance of the patient to ensure the setting is prepared. In total, the therapist pair will ensure the room is clean, airy and comfortable; that blankets, pillows, and sheets are on the bed; that an eye mask and headphones are available; that lighting is dim and comfortable; that all audio equipment is tested and is working (including headphones); that all medical equipment is available and working (BP
machine, thermometer); that medication is collected from a pharmacy/stored on-site, and appropriate access has been organized; that rescue medication is accessible;
and that therapists' needs are taken care of.
[395] The therapist pair will then conduct a pre-dosing checklist upon patient arrival, which includes reconfirming consent; obtaining blood pressure and pulse measurements; assessing suicide risk via C-SSRS; conducting a urine screen (for substances and pregnancy as appropriate), conducting an alcohol breath test; checking participant medications, (including any changes and what, if any, medication the patient has taken that day to ensure that, e.g., if hypertensive medication was taken that such is routine, etc.).
[396] The patient will then be reminded of key information, including that they are expected to remain in the facility (and in the room) for at least 6 hours after the initial dose, that vital signs will be monitored at dosing, 1 hour after the initial dose, 2 hours after the initial dose (before offering the booster dose), 3 hours after the initial dose, and 6 hours after the initial dose (at the end of the session). That the patient can be supported to get to the bathroom as needed, and the patient is subsequently orientated to the default position of laying on the bed, while wearing eye shades and headphones. That the patient should embrace the experience as much as possible, to be open and curious to all experiences that airse. That the patient will never be on their own, and that at least one therapist will be with them at all times. The patient is also reminded about how to reach out and/or ask for support, and the intention set for the session is reviewed. Finally, the therapist pair asks for any questions, reflections, comments, or concerns the patient may wish to talk about at this point.
[397] During the drug-assisted session, the therapeutic interaction will benefit from being more patient-led and open, with the therapist pair maintaining a present and interested stance;
holding the PFM in mind but refraining from delving deeply into these processes during the drug session (this can occur during subsequent integration).
[398] When assessing vitals and recording measurements, the therapist pair may gently inquire about how the participant is doing, and if they have been largely 'inside' during the session. A gentle touch on the arm (if they have consented to touch) and a question such as, 'just checking in how you're doing' will usually be sufficient. Another point at which the participant may naturally wish to engage may come when the booster dose is offered. Here, the therapist pair may wish to inquire more about how the participant is progressing and where they currently find themselves. All communication is offered as a suggestion or invitation rather than an instruction. For example, 'would you like to. . 'might this be a good time to...' etc. Occasional interactions will help the therapist team judge whether some additional interaction may assist the therapeutic process, whilst always being respectful of the participant's own healing intelligence. This acknowledges and trusts that the participant has an innate internal ability to heal themselves, with the therapy used as a method to facilitate this process and support its natural unfolding. The therapists can use the therapeutic formulation to guide them as to how much support may be required to facilitate the participant's processing.
For example, if the participant has identified a tendency to cope via shutting down or isolating themselves, a reminder of the therapist pair's presence and availability may be useful.
However, if a participant may intellectualize as an avoidance strategy, they may be encouraged to spend more time 'inside.'
[399] The therapist pair may also inquire about any sensations in the body associated with what the patient is describing, which can also be helpful to ensure the patient stays focused on their internal experience. Additionally, asking about imagery can support an internal focus. For example, 'when you describe feeling afraid, do you notice any sensations in your body?' and 'are there any images in your mind when you talk about that?'
[400] If the participant describes intense challenging emotions, the therapist pair may offer verbal support and/or supportive touch. They may say, 'see if you can stay with that feeling that's coming up for healing as much as you can, we're here with you'.
[401] As the majority of actions undertaken by the therapist pair during a dosing session are to support the participant's internal processing and to stay with internal experiences that arise, the therapist pair will be acutely aware of any signs that the patient is unable to maintain an internal focus. When such arises, the therapist pair may intervene with supportive actions, such as but not limited to reminding the patient of their presence e.g., 'we are here with you', you are safe,' using supportive touch (if pre-agreed), encouraging the use of breath, 'breathe into it,' remember to use your breathing,' etc.
[402] This instruction can become more directive as warranted (e.g., in accordance with levels of distress). For example, 'breathe with me' or 'breathe in and out on my count' may be used.
[403] If the patient's levels of distress do not appear to be manageable over a prolonged period, the therapist team can call the study medic who will then make an assessment and consider whether the option of rescue medication is appropriate. In such cases, up to 4 mg of Lorazepam may be administered over a single visit (i e , one session)
[404] Towards the end of the dosing session, the therapist may make general inquiries about how the patient coped during the session, and how they are presently feeling.
However, it is best to not press the patient for a full narrative of the experience at this time, as there may be on-going processing and/or the patient may wish to be quiet and have a period of reflection and introspection. If the patient does wish to talk about their experience at this point, the therapist may listen as a supportive presence but limit any interpretation at this stage while the patient may be in a phase of increased psychological flexibility, and processing may still be unfolding.
Rather, it is recommended the therapist pair encourage the patient to have a quiet rest of the day, and to journal/make notes of the experience. Note, the patient may also wish to express their feelings in other ways, such as but not limited to art or music.

Week 3, Session 4
[405] The first session after a dosing session will be geared towards integrating the MDMA
experience with the prior therapy sessions. The therapist pair will discuss the purpose of the MDMA dosing session, and will have prompt questions for the patient. At this stage, the therapist pair will also introduce mindfulness techniques and practice the same.
[406] Broadly, mindfulness will be introduced as a key element of 'being present,' and is a technique useful in paying attention to the present moment non-judgmentally.
It is often a skill taught as part of relapse prevention, and allows patients to notice their "present moment." It will be useful to ask if the participant has any previous experience practicing mindfulness, and how such experiences went This session will end with a mindful "body scan"
which will be readily known in the art.
Week 4, Session 5
[407] During the fifth therapy session, the therapist team will continue previous discussions regarding processing the material that emerged during the MDMA dosing session.
In addition, High Risk Situations (FIRS) and coping responses for the same, will be introduced and discussed.
Week 5, Session 6
[408] During the sixth therapy session, the therapist team will discuss the practice of "unhooking from one's thoughts,- and will prepare the patient for the second MDMA dosing session.
Week 6, Sessions 7 & 8
[409] During the seventh and eighth therapy sessions, the therapist team will ensure the patient is ready for their second experiential MDMA dosing session, and will lead journaling and mindfulness practice.
Week 7, Session 9
[410] During the ninth therapy session, the therapist team will continue to discuss material that emerged during the MDMA dosing session. The therapist team will also discuss the concept of "opening up by letting go of the struggle."
Week 8, Session 10
[411] During the tenth therapy session, the therapist team will discuss what matters to the patient, including but not limited to key personal values, and will assist the patient in goal setting in accordance with said values. Finally, the therapist team -Identifying barriers and responses Week 9, Sessions 11 & 12
[412] During the eleventh and twelfth therapy sessions, the therapist team will ensure the patient is prepared for their third MDMA dosing session, and will lead journaling and mindfulness practice.
Week 10, Session 13
[413] During the thirteenth therapy session, the therapist team will instruct the patient how to respond to urges to drink, will aid the patient in "stepping out of automatic pilot," (i.e., being more present), and will discuss "Seemingly Irrelevant Decisions," (SIDS), which are a series of decisions made that, while appearing to be innocuous, lead a patient to a situation wherein alcohol may be freely consumed; "responding to lapses and relapses,"
"developing self-compassion," and how to develop a "SOBER breathing space" with the patient.
[414] Broadly, a "SOBER breathing space" refers to an acronym useful in diagnosing, and responding to dangerous/tempting situations, wherein "S" refers to "stop," so the patient is instructed to stop at a given "challenging point," and make the choice to step out of automatic pilot. "0," stands for "observe," wherein the patient must become aware of what is happening within themselves. This includes appropriately diagnosing physical sensations, emotions, and, generally, what is happening within the body. This will enable the patient to notice any urges that may force them to act in a certain way. The patient must not force anything away or keep anything out, but instead acknowledge what is happening. "B," stands for "breath," wherein the patient is instructed to gather their intention and focus on their breathing. The anchors of the breath are used to come into the body and to stay present. "E," stands for "expand," wherein the patient expands the awareness they now feel to include a sense of the entire body, heart, and mind _________ including any tightness or tension, present emotions, etc.
Finally, "R" stands for "respond," wherein the patient applies the awareness they now feel to their current situation, to determine if the emotion, setting, or person the patient is with is dangerous for them and their recovery. The patient examines the choices they have, and is aware that they are in control of what choice they ultimately make. They then choose the option that best fits what the patient wants in life, including who they wish to be, and what is truly important to them. The patient then visualizes making that choice, and follows through with the decision.
Week 11, Session 14
[415] During the fourteenth therapy session, the therapist team will review the therapy course with the patient.
[416] It is at this time the therapist pair is reminded to engage in self-care. This work can be extremely rewarding and also intense and tiring. It is essential that therapists look after themselves. This will help them to be as available and supportive to patients as possible. The therapist pair is encouraged to take some time to consider what the potential challenges of this work may be for them. There are likely to be some practical and personal considerations that can be helpful to discuss with their supervisor early on.
[417] Broadly, when working with a co-therapist, it is important to have met or spoken with them before the patient session (this could be in person, over the telephone or via video call).
This will give the opportunity to establish rapport together and also to consider practical considerations such as how to divide the work. It is important to feel comfortable to proceed with the patient and to discuss with the allocated supervisor if there are any doubts..
EXAMPLE 8: Open-Label, Within-Subjects, Safety and Tolerability Feasibility Study of MDMA-Assisted Psychotherapy in Patients with AUD
[418] In this Example is described the first study to investigate the use of MDMA to treat AUD: an open-label safety and tolerability proof-of-concept study investigating the potential role for MDMA therapy in treating patients with AUD. The study was designed to assess if MDMA-assisted psychotherapy can be delivered safely and can be tolerated by patients with AUD post-detoxification. Outcomes regarding abstinence from alcohol, quality of life and psychosocial functioning also were evaluated. Before this study, the qualities of MDMA
therapy that could make it potentially well-suited for patients with addictions had been raised in theory by inventor hereof (Sessa 2017), and queried ("Will MDMA work for addictions?"), but they had never before been explored in a research study.
[419] In the study of this Example, 14 patients with AUD completed a community alcohol detoxification course and received an eight week course of abstinence-based therapy, such as described above. Participants received two sessions with MDMA (187.5 mg each session, as two separate doses of 125 mg and then 62.5 mg). Psychological support was provided before, during, and after each session. Safety and tolerability were assessed alongside psychological and physiological outcome measures. Alcohol use behavior, mental well-being and functioning data were collected for nine months after alcohol detoxification.
[420] The study of this Example demonstrated that MDMA treatment was well-tolerated by all participants. No unexpected adverse events were observed. Psychosocial functioning improved across the cohort. Regarding alcohol use, at nine months post-detox the average units of alcohol consumption by participants was 18.7 units/week, compared with 130.6 unit/week pre-detox. This compares favorably to a previous observational study by the same team with a similar population of people with AUD (Sessa et al. 2020, and reproduced as FIGS. 1 and 2).
For the first time, and in numerous unexpected and surprising ways, support for the safety and tolerability of this novel intervention for AUD post-detox was shown.
[421] Specifically, as see in FIG. 1, all but one participant was successful in remaining abstinent from alcohol for at least one month after detox (after averaging 196.7 units of alcohol per week pre-detox) and, while the consumption of alcohol increased from one month to 3 months (from an average of 5.2 units of alcohol consumed per week to an average of 120.1 units of alcohol consumed per week), the figure gradually decreased from 3 months to 7 months, reaching a low of 106.9 units of alcohol consumed per week, and ending at 113.9 units of alcohol consumed per week (As noted in FIG. 2).
[422] Such results, in view of the further description and disclosures provided herein, demonstrate for the first time the therapeutic potential of MDMA-assisted psychotherapy for this population of people with physically dependent AUD, who required and underwent detoxification.
[423] This Example details an open-label, within-subjects, safety and tolerability feasibility study that was conducted in 14 patients aged 18-65 years old diagnosed with AUD who had recently undergone detoxification, and received MDMA-assisted therapy. The main outcome measures of this study were the number of patients completing the eight-week course, the number accepting the second booster dose of MDMA on drug-assisted days, and adverse events. Secondary outcome measures included changes in drinking behavior (measured by units per week consumed at 3, 6 and 9 months since completion of detoxification), measures of mental well-being, psychosocial functioning, quality of life, and concomitant drug use.
Study Design
[424] Patients with a primary diagnosis of AUD who were seeking detoxification¨with or without medical assistance¨were recruited from the North Somerset Substance Misuse Service (Addaction). Patients received an eight-week course of recovery-based therapy comprising 10 psychotherapy sessions, two of which included dosing patients with open-label MDMA during a 6 to 8-hour assisted therapy session. For each dosing session, participants received an initial oral dose of about 125 mg MDMA, followed about 120 minutes later by a booster dose of about 62.5 mg MDMA, in which the booster dose served to prolong the experience, which allowed for greater time for psychotherapy under the influence of the drug.
[425] Other sessions, including sessions 1, 2, 4, 5, 6, 8, 9, and 10, comprised one-hour psychotherapy sessions, wherein aspects of motivational interviewing and "third-wave"
cognitive-behavioral approaches were employed. In all, patients remained in the study for approximately 10 months.
Inclusion and Exclusion Criteria
[426] Inclusion criteria and exclusion criteria included those as set forth in TABLE 1. If one of the exclusion criteria were met, an individual was not selected to take part in the study. To be considered as a patient, all individuals also had to meet all inclusion criteria.
Methods
[427] AUD was identified using the DSM-IV SCID interview. Screening comprised of written informed consent, an evaluation of the patient's physical and mental health background, a psychiatric interview (MINI), and assessments of depression and anxiety severity using the Patient Health Questionnaire-9 (PHQ-9) and Generalised Anxiety Disorder Assessment (GAD-7) questionnaires. Severity of AUD was established using the Severity of Alcohol Questionnaire (SADQ) and the Short Inventory of Problems for Alcohol (SIP) questionnaire.
However, it should be noted, additional questionnaires/assessments equivalent to those mentioned (as would be apparent to one of skill) may also be used. Patients received a thorough physical health check comprising an electrocardiogram, routine blood tests, blood pressure, heart rate and physical examination. Following screening, eligible patients underwent the process of detoxification either by gradually cutting down alcohol consumption over many weeks or with a medically assisted detoxification regime. The majority of participants were also taking medications for anxiety and/or depressive symptoms (e.g. selective serotonin reuptake inhibitors). According to the inclusion/exclusion criteria, associated medications known to attenuate the effects of MDMA were subsequently gradually reduced and stopped under medical supervision ahead of the first MDMA session. A further 'baseline' visit clarified successful detoxification using the Clinical Institute Withdrawal Assessment for Alcohol (CIWA) questionnaire before eligible participants entered the eight-week course of psychotherapy. This entailed weekly 60-minute outpatient non-drug psychotherapy sessions delivered by two clinicians (B.S. and L.H.) trained in delivering MDMA-assisted psychotherapy by the USA-based organisation MAPS.
[428] Dosing with MDMA occurred twice during the eight-week course on weeks 3 and 6.
Physiological changes, observer and subject ratings of distress (Subjective Units of Distress (SUDS)) and the intensity of MDMA's acute psychoactive effects were measured throughout the drug-assisted session_ Acute anxiety was managed primarily psychologically, but sedative medication (oral lorazepam) was available. Participants remained overnight in the treatment center after each drug-assisted session, overseen by medically trained 'night sitters' who were on hand to support participants as required but instructed to avoid delivering any psychotherapeutic interventions.
[429] Participants were seen the morning after each drug-assisted session for an integration psychotherapy session, and then telephoned daily for six days to assess changes to mood, suicidal risk factors (using the C-SSRS) and quality of sleep (using the Leeds Sleep Evaluation Questionnaire). Following the end of the eight-week therapeutic course, participants carried out additional follow-up questionnaires. They were then seen again at three, six, and nine months (since baseline) for longer-term follow-up data collection.
Data Analysis
[430] All data were recorded on paper case report forms and then digitized into MS Excel spreadsheets. Analysis and graphing were performed with GraphPad Prism v.8.4.3 (GraphPad Software LLC, La Jolla, CA) or MS Excel. When calculating timeline follow-back results, alcohol consumption levels at last observation were used in the case of drop-outs or when participants had undertaken a second detoxification (Hamer and Simpson, 2009).
Results
[431] Demographics: Thirty-six participants attended face-to-face screening visits, and 14 were enrolled (8 males and 6 females; the mean age being 48 years). All were white British.
Four were employed, nine were unemployed and one was retired. The average age of first alcohol use was 13 years old. The average age when alcohol became problematic was 34 years old. Nearly two-thirds (64%) of participants reported a history of alcohol-related blackouts, 14% had experienced alcohol withdrawal¨induced seizures, 86% of participants reported having experienced risky or vulnerable incidences due to alcohol and 75% of participants had had forensic/offending behaviour secondary to their alcohol use.
[432] Severity of AUD criteria at screening and baseline: As per the inclusion criteria, all eligible patients scored above the diagnostic threshold on the DSM-5 SCID
questionnaire for AUD. AUD severity was also measured using the SIP questionnaire and the SADQ
questionnaire (see FIG. 6), with most eligible participants in the moderate to severe range. At the point of Baseline visit, (within one week of detox completion), 100% of eligible participants had successfully completed detoxification, assessed using the CIWA scale.
[433] Physiological and tolerability effects during MDMA sessions: In total, 26 drug-assisted psychotherapy sessions with MDMA were administered during the trial, Temperature, blood pressure and heart rate were measured at t=0, before taking MDMA, then half-hourly up to t=2 hours, then hourly thereafter for a minimum of 6-hours from the time of dosing (see FIG. 7).
[434] Except for one participant, all of these physiological parameters remained within normal limits for all these sessions. As expected, a mild, transient rise in blood pressure, temperature and heart rate over the course of the MDMA session was seen. No patients experienced sustained abnormal physiological disturbance, symptomatic experiences of raised blood pressure, heart rate or temperature or any other adverse events during MDMA sessions.
No medical interventions were required in respect of these or any other physiological events during MDMA sessions.
[435] One participant experienced a transient abnormal rise in blood pressure after taking the initial dose of 125 mg MDMA reaching a BP 183/118 at two hours after dosing, attributed to the participant forgetting to take her regular antihypertensive medication on the morning of dosing. Although she was asymptomatic and no medical intervention was required it was decided to withhold the two hour supplemental dose. Her blood pressure subsequently spontaneously returned to normal in the following two hours.
[436] Subjective Units of Distress (SUDS) and Participant report of Drug Effects were also measured, hourly, throughout the MDMA sessions (see FIG. 7). Most subjects predictably reported mildly raised SUDS scores at the beginning of the sessions before taking MDMA¨
consistent with expected anxiety ahead of dosing¨which subsequently reduced during the course of the session as the positive effects of MDMA emerged. Participants gave their own subjective score (0-10) of whether they felt Drug Effects, and the therapists also recorded their own obj ective score of how "altered- the participant appeared. There was no significant difference between Observers' and Participants' Drug Effects scores. Drug Effects rose expectedly over the first two hours, with a notable further increase after the booster dose was given at t=2 hours, and a subsequent plateau then decline over the following six hours. By the end of the MDMA session day all drug effects had returned to baseline. No participants reported any significant neurocognitive impairments associated with receiving MDMA in the weeks and months following participation in the study.
[437] Changes in drinking behavior: While changes in drinking behaviour were not a primary outcome measure, we nevertheless collected data in respect of units of alcohol consumed per week (wherein a standard drink contains about 14 g of pure alcohol, i.e., ethanol) in the month before participants' detoxification, immediately after detox (baseline'), throughout the eight-week MDMA therapy course and for up to nine months after detox. Of the 14 eligible participants who underwent the course of MDMA-assisted psychotherapy, at the nine-month follow-up end point, 11 participants were drinking fewer than 14 units of alcohol per week (including nine who were totally abstinent from alcohol), and three participants had relapsed to drinking more than 14 units of alcohol per week.
On average, participants were drinking 130.6 units of alcohol per week in the month before detoxification, and no units at the point of detox. After nine months, the average amount of consumed alcohol had risen back to 18.7 units per week (see FIG. 8).
[438] Specifically, participants consumed a cumulative 16.9 units of alcohol per week at 3 months post-detox, 38.0 units of alcohol per week at 6 months post-detox, and 18.7 units of alcohol at 9 months post-detox. This yields decreases of 87.1% at 3 months post-detox, 71% at 6 months post-detox, and 85.7% at 9 months post-detox.
[439] Seven-day followup after MDMA sessions: Considerable medical literature and the popular press report the anecdotal observation of ecstasy users experiencing an acute 'come-down' effect and a drop in mood in the days after using the drug recreationally. In order to measure this prospectively with clinical MDMA, we measured participants' mood states by daily Profile Of Mood States (POMS) measurements for seven days after each MDMA session (see FIG. 9). Positive scores represent depressed affect, zero represents no change in mood/affect and results below zero represent a positively felt mood. Average scores across both MDMA sessions for all 14 participants (261VIDMA sessions) revealed no evidence of any mood disturbance during the week after taking each session of clinical MDMA.
Indeed, participants sustained a positive mood for seven days. This result contrasts with anecdotal reports from recreational ecstasy users.
[440] Other mental health measures and quality of life measures: Brief assessments of mood and anxiety were made at screening, baseline, after the eight-week MDMA
therapy course and at the three-, six-, and nine-month follow-ups using the PHQ-9 and GAD-7 rating scales, respectively (see FIG. 10). Scores demonstrate a reduction in both anxiety and depression after screening and baseline time points, followed by a transient rise in anxiety and depression scores three months after baseline, a further reduction at six months and a moderate rise again at nine months post detox. A variety of further data were collected, including changes to the quality of sleep, quality of life measures and changes to compassion and empathy scales.
These data will show improvements in all such secondary measures.
[441] Suieidality: Participants underwent the C-SSRS at screening, baseline, throughout the eight-week therapy course, in the week after each MDMA session and at the three-, six-, and nine-month follow-up visits. No participants reported current suicidal ideation, intent or plans or self-harm behaviour during the course of the study.
[442] Adverse events: The acute effects of MDMA-assisted psychotherapy were well tolerated by participants. No unexpected adverse events occurred. No participants reported any desire to use illicit ecstasy/illicit MDMA following receiving clinical MDMA
as part of this trial. No psychotic symptoms were observed in any of the patients.
[443] A variety of further data were collected, including changes to the quality of sleep, quality of life measures and changes to corn- passion and empathy scales, which will be published in forth- coming papers.
Discussion
[444] In this first safety and tolerability study, MDMA-assisted psychotherapy was demonstrated for the first time to be useful in treating AUD, for example in patients needing detox and physically dependent on alcohol.
[445] Prior to carrying out the study of this Example, the same study team carried out a non-interventional observational study, following 14 participants through their treatment-as-usual post-alcohol detox (the "Outcomes Study," published as Sessa et al., 2020).
The eligibility criteria and questionnaires used in the Outcomes Study had similarities to the study of this Example, but it was without the additional eight-week therapeutic course with MDMA-assisted psychotherapy, which occurred post detox. Although patients were not randomized into the studies, FIG. 11 demonstrates the success of the participants of the study of this Example in terms of alcohol consumption over 9 months, compared with current best treatments available locally. Only 21% of participants who had undergone MDMA-assisted psychotherapy were drinking in excess of 14 units of alcohol a week in comparison with the 75%
observed in the Outcomes Study (see FIG. 11).
[446] Through the Example, MDMA-assisted psychotherapy is demonstrated to be useful in treating AID, and in AID in patients who are physically dependent on alcohol EXAMPLE 9: Randomized Double-Blind Between-Subject Controlled Study of MDMA-Assisted Psychotherapy in Patients )4,ith "Dependent Use" AUD
[447] In this Example is described a randomized, double-blind controlled study of efficacy of MDMA-Assisted Psychotherapy for the treatment of patients with physically dependent AUD
("Dependent Use"). This is a randomized between-subject controlled study, in patients with physically dependent AUD. Half of the patients will receive MDMA-Assisted psychotherapy;
the other half will receive placebo-assisted psychotherapy.
[448] In this study it will be shown that MDMA-assisted psychotherapy is an effective treatment for patients with AUD, who have physical dependence. In other words, the study of this Example will show that MDMA-assisted psychotherapy is superior to placebo dose-assisted psychotherapy and can reduce use of alcohol in patients with physical alcohol dependence¨and, in some embodiments, especially those who require a course of detoxification before they undergo drug-assisted therapy ________________________ and improve their quality of life.
[449] Outcomes regarding abstinence from alcohol, concomitant drug use (the number and frequency of both prescribed medication use and recreational drug use, including any craving related to MDMA and subsequent use of illicit ecstasy), quality of life, and psychosocial functioning will be evaluated. The main outcome measure will be efficacy, which will be measured in weekly units of alcohol consumed, (wherein a standard drink contains about 14 g of pure alcohol, i.e., ethanol) at 3, 6 and 9 months post detox.
[450] Here, as also in the Examples above, the study will adhere to the principles outlined in the Medicines for Human Use (Clinical Trials) Regulations 2004 (SI 2004/1031), amended regulations (SI 2006/1928) and the International Conference on Harmonisation Good Clinical Practice (ICH GCP) guidelines, the Data Protection Act, and such other regulatory requirements as appropriate and as would be understood to those of ordinary skill in the art.
[451] A population of adult patients with a primary diagnosis of AUD, aged between 18 and 65, who have physical dependence (and therefore in some embodiments will require, and undergo, medical detoxification) and who meet the inclusion and exclusion criteria, will be recruited (in a number sufficient to generate statistically significant results). Patients will remain in the study for a duration of approximately 14 months.
[452] As depicted in the table of FIG. 17 (hut with such changes as will he readily appreciated, such as the replacement of "Pre-Therapy Prep-Group" with pre-detox group therapy, followed by detoxification, compare, e.g., TABLE 4 and TABLE 8), and as also further described in the other Examples herein, patients will first receive four weekly sessions of motivational or supportive group therapy pre-detox. Subsequently, alcohol detoxification will be achieved either by gradual reductions in daily alcohol use (which may begin from the start of the pre-detox group phase, with support from the group facilitators) and/or through a medical detox. Once abstinent, they will enter a 13-week course of MDMA-assisted therapy, comprising fifteen psychotherapy sessions (or, in an alternate aspect, an eight-week course as in the embodiment described above). Patients are randomized to either subtherapeutic / active control (placebo) or therapeutic dose. On three psychotherapy sessions (session 3, session 7 and session 13) patients will be given either active control (20 mg + 10 mg subtherapeutic) dose of MDMA or active therapeutic (125 mg + 62.5 mg) dose MDMA during the 6-8 hour assisted therapy sessions. Patients may optionally stay overnight, or go home after their MDMA session. Other sessions (1, 2, 4-6, 8-12, 14 and 15) will comprise non-drug-assisted 1-hour psychotherapy sessions.
[453] The sessions in general will be as described above, and reference is thus made to the description in the above Examples. From Session 15 or if discontinued treatment, from 2 weeks post the final psychotherapy session to the final follow-up visit (9 months post baseline).
Patients will be followed up at 3, 6, and 9 months from the date of baseline.
[454] Results are obtained as above, and through this Example, MDMA-assisted psychotherapy is further demonstrated to be useful in treating AUD, and in particular AUD in patients who are physically dependent on alcohol, and to have such other benefits as claimed.
[455] Accordingly, and in view of all of the Examples herein, in some embodiments, the pharmaceutical compositions of the invention (and their use in the methods of the invention) are used to improve the symptoms of a mental health disorder, wherein the mental health disorder is a substance abuse disorder. In some embodiments, that substance use disorder is AUD, and specifically physically dependent alcohol addiction. The symptoms of the mental health disorder to be treated (e.g., the symptoms of AUD or physically dependent alcohol addiction) shall be able to be determined by one of skill, by reference to the general understanding of the art regarding that disorder, for example by reference to the DSM-5).
Accordingly, methods of the invention, and the compositions of the invention when used in those methods, will be understood as improving any such symptoms.
[456] In some embodiments, the pharmaceutical compositions of the invention (and their use in the methods of the invention) will produce a therapeutic effect, and such effect may be (1) an improvement in a symptom of a comorbid psychiatric disorder; (2) an increase in quality of life; (3) an increase in psychosocial functioning; (4) a decrease in use or frequency of prescription medication, (5) a decrease in use or frequency of recreational drugs, which can include illicit use of prescription drugs; (6) a decrease in units of alcohol consumed; (7) a reduction of cravings relating to alcohol; (8) a prevention of relapse.
E. Therapeutic Outcomes
[457] In some embodiments, the methods of the invention, such as in the eight-week or 13-week MDMA-assisted psychotherapy course, or other embodiments disclosed herein, will provide one or more therapeutic effects for a patient or a subject diagnosed with or having AUD, or symptoms thereof, or otherwise in need of such treatment. In some embodiments, the methods of the invention will result in one or more of: (a) a reduction of alcohol use, (b) a reduction of alcohol cravings, (c) a prom oti on of al cohol abstinence, (d) a preve-nti on of relapse into alcohol use, or (e) an improvement of at least one symptom of alcohol use disorder. In some embodiments, the methods of the invention will result in one or more of:
(a) an increase in quality of life, (b) an increase in psychosocial functioning, (c) a decrease in use or frequency of a prescription medication, (d) a decrease in use or frequency of a recreational drug, (e) a decrease in obsessive compulsive thoughts, (f) a decrease in suicidality, (g) an increase in feelings of empathy, or (h) an increase in self-compassion. In some embodiments, the methods of the invention will result in an improvement of at least one symptom of a comorbid psychiatric disorder, such as antisocial personality disorder, borderline personality disorder, depression, anxiety, schizophrenia, attention deficit hyperactivity disorder, bipolar disorder, obsessive compulsive disorder, binge eating disorder, or PTSD.
[458] In some embodiments, the methods of the invention, such as in the eight-week or 13-week MDMA-assisted psychotherapy course, or other embodiments disclosed herein, will provide one or more therapeutic effects for a patient or a subject diagnosed with or having AUD, or symptoms thereof, or otherwise in need of such treatment, and will result in one or more of: (a) an increase in physical functioning; (b) a decrease in role limitations due to physical health; (c) an increase in emotional well-being; (d) a decrease in role limitations due to emotional problems; (e) an increase in energy; (f) a decrease in fatigue;
(g) an increase in social functioning; (h) a decrease in pain; or (i) an increase in general health. In some embodiments the increase and/or the decrease will be demonstrated by a patient-reported measure on a Short Form (36) Health Survey, or as otherwise described in Sessa (2020) and shown here, for example, in FIG. 5 and accompanying description thereof.
[459] In some embodiments, the measurement of such therapeutic effects may be performed by a therapist or other clinician (e.g., an interview), by a patient his or herself (e.g., a self-reported questionnaire), by a third-party human or by a medical or other device (e.g., a medical sensor or biosensor, a watch or fitness tracker); such measurements may be graded by a human decision-maker or an AT, by using machine learning, or by a computer algorithm.
[460] In some embodiments, any one or more of such therapeutic effects will be durable effects, which will be shown when measured at a time following treatment by at least one month, at least three months, at least six months, at least nine months, at least one year, or greater than one year, including when measured at times in between. Such measurements "following treatment" may be measured, e.g., from the end of detoxification, if detoxification is completed, from baseline, from intake, or from the first psychotherapy s es si on, and therefore "following treatment" may include a period measured from the start of treatment, or a point following the start of treatment, but before treatment is complete In other embodiments, a measurement is from the conclusion of a therapy protocol, or after the conclusion of a certain or chosen phase of a therapy protocol. In other embodiments, a measurement is from the end of treatment, i.e., after treatment is complete, which in some embodiments will be from the completion of the MDMA-assi sted psychotherapy regimen.
[461] In some embodiments, the methods of the invention will result in a reduction of alcohol use. In some embodiments, a reduction of alcohol use may be measured by a reduction in the units of alcohol consumed per week (compared to the units of alcohol consumed per week prior to undergoing the methods of the invention), wherein a unit of alcohol refers to a standard drink, which contains about 14 g of pure alcohol (ethanol). For typical alcoholic beverages, this refers to 12 ounces of 5% alcohol beer, 5 ounces of 12% wine, and 1.5 ounces of 40%

distilled spirits (NIH). In some such embodiments, the amount of alcohol consumed per patient is a durable effect, and will be shown at least 1 month, at least 3 months, at least 6 months, at least 9 months, or at least 1 year following treatment. In some embodiments, the amount of alcohol consumed per patient is reduced by about 5% to about 50%, including about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, and values in between. In some preferred embodiments, the amount of alcohol consumed per patient is reduced by 50% or more. In some preferred embodiments, the amount of alcohol consumed per patient is reduced by at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, up to and including by 100% (i.e., a patient is no longer consuming alcohol), and values in between.
[462] In some embodiments, the methods of the invention will result in a reduction in alcohol cravings. In some embodiments, a reduction in alcohol cravings may be measured by subjective patient reports, as well as questionnaires, such as but not limited to the alcohol craving experience questionnaire. In some such embodiments, the reduction in alcohol cravings is a durable effect, and will be shown at least 1 month, at least 3 months, at least 6 months, at least 9 months, or at least 1 year following treatment. In some embodiments, alcohol cravings will be reduced by about 5% to about 100%. In some embodiments, alcohol cravings will be reduced by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, up to and including by 100% (i.e., the patient is no longer having alcohol cravings), and values in between.
[463] In some embodiments, the methods of the invention will result in a promotion of abstinence, wherein abstinence refers to a prolonged period of time-such as at least 1 day, at least 2 days, at least 3, at least 4 days, at least 5, at least 6 days, at least 7 days, or more than 7 days-wherein the patient does not consume alcohol. In some embodiments, a promotion of abstinence may be measured by measuring the average duration of abstinence (e.g., the average time between "heavy drinking days," as defined herein), before undergoing the methods of the invention, and after undergoing the methods of the invention. In some such embodiments, the promotion of abstinence is a durable effect, and will be shown at least 1 month, at least 3 months, at least 6 months, at least 9 months, or at least 1 year following treatment. In some embodiments, the promotion of abstinence will be illustrated by an average increase in time between "heavy drinking days," as defined herein, by at least about 5%, preferably by at least 25%, more preferably by at least 50%, and most preferably by at least 100%. In some embodiments, the promotion of abstinence will be illustrated by an average increase in time between "heavy drinking days," as defined herein, by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, and about 100%, and values in between. In some most preferred embodiments, the promotion of abstinence will be illustrated by an average increase in time between "heavy drinking days," as defined herein, by at least 100%, such as at least about 150%, at least about 200%, at least about 250%, at least about 300%, at least about 350%, at least about 400%, at least about 450%, at least about 500%, at least about 550%, at least about 600%, at least about 650%, at least about 700%, at least about 750%, at least about 800%, at least about 850%, at least about 900%, at least about 950%, at least about 1000%, and, in some embodiments, the promotion of abstinence will be illustrated by an average increase in time between "heavy drinking days," as defined herein, by more than about 1000%, as well as values in between.
[464] In some embodiments, the methods of the invention will result in a prevention of relapse into alcohol use. In some embodiments, a prevention of relapse into alcohol use may be assessed by measuring the amount of alcohol consumed after undergoing the methods of the invention, and identifying whether or not the amount of alcohol consumed-and frequency with which it is consumed-is above the threshold to qualify the person as a "heavy drinker,"
as defined herein, wherein a "heavy drinker" refers to an individual who "binge drinks" at least or more days in a given month, and "binge drinking" refers to a male consuming 5 or more alcoholic drinks on the same occasion, or a female consuming 4 or more drinks on the same occasion. In some such embodiments, the prevention of relapse into alcohol use is a durable effect, and will be shown at least 1 month, at least 3 months, at least 6 months, at least 9 months, or at least 1 year following treatment. In some embodiments, a male patient will consume 5 or more drinks on the same occasion less than 5 times in a given month, for at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, at least 6 months, at least 7 months, at least 8 months, at least 9 months, at least 10 months, at least 11 months, or at least 12 months following the completion of the methods of the invention. In some embodiments, a female patient will consume 4 or more drinks on the same occasion less than 5 times in a given month, for at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, at least 6 months, at least 7 months, at least 8 months, at least 9 months, at least 10 months, at least 11 months, or at least 12 months following the completion of the methods of the invention. In some embodiments, a prevention of relapse into alcohol use will be a reduction in the rate of relapse or likelihood of relapse, for example, a reduction of about 5% to about 100%. In some embodiments, a reduction in rate or likelihood of relapse will be about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, up to and including 100% (i.e., complete prevention of relapse), and values in between.
[465] In some embodiments, the methods of the invention will result in an increase in quality of life. In some embodiments, an increase in quality of life may be measured by subjective reports from patients, questionnaires, such as, but not limited to, the Quality of Life Scale (QoLS), the Health Related Quality of Life (HRQoL) questionnaire, World Health Organization Quality of Life instruments, for example, WHOQOL-100 and its abbreviated version, WHOQOL-BREF, the Munich Quality-of-life Dimension List (MLDL), Medical Outcomes Study Short Form (SF-36), the Quality of Life Enjoyment and Satisfaction Questionnaire (Q-LES-Q), the EuroQoL-5D (EQ-5D), the Life Situation Survey (LSS), the Nottingham Health Profile (NHP), the Sleep Quality Scale (SQS), the Pittsburgh Sleep Quality Index (PSQI), and the Leeds Sleep Evaluation Questionnaire (LSEQ), as well as changes in mental health conditions, including but not limited to anxiety and depression (i.e., after completing the methods of the invention, confirming the severity of the patient's anxiety and/or depression has decreased compared to the severity of the same prior to undergoing the methods of the invention). In some embodiments, anxiety and/or depression are assessed using questionnaires, for example, the Patient Health Questionnaire (PHQ-9) or the Generalized Anxiety Disorder Assessment (GAD-7). In some such embodiments, the increase in the patient's quality of life is a durable effect, and will be shown at least 1 month, at least 3 months, at least 6 months, at least 9 months, or at least 1 year following treatment.
In some embodiments, the patient's quality of life will be increased by between about 5%, to about 100%. In some embodiments, the patient's quality of life will be increased by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100%, and value in between. In some embodiments, wherein the patient's quality of life is determined by a reduction in depression, the patient's depression may decrease by between about 5%, to about 100%. In some embodiments, wherein the patient's quality of life is determined by a reduction in depression, the patient's depression may decrease by between about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100%, and values in between (i.e., the patient no longer has quantifiable depression). In some embodiments, wherein the patient's quality of life is determined by a reduction in anxiety, the patient's anxiety may decrease by between about 5%, to about 100%. In some embodiments, wherein the patient's quality of life is determined by a reduction in anxiety, the patient's anxiety may decrease by between about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, up to and including 100% (i.e., the patient no longer has quantifiable anxiety), and values in between.
[466] As highlighted in FIG. 3, the methods of the invention are effective in reducing mild, moderate, moderately severe, and severe depression. Prior to undergoing the methods of the invention, specifically during the screening session, 12 patients were assessed for levels of depression. It was found that one patient suffered from mild depression, two suffered from moderate depression, four suffered from moderately severe depression, and five suffered from severe depression. Directly after going through the detoxification process (noted as "baseline"
in FIG. 3), one patient had mild depression, two had moderate depression, zero had moderately severe depression, and two had severe depression (out of 11 patients remaining in the study).
The patients were then reassessed at 3, 6, and 9 months post-baseline (i.e., after completing a detoxification course, and prior to beginning the MDMA-assisted psychotherapy regimen), wherein it was discovered that, at 3 months, two had mild depression, one had moderate depression, three had moderately severe depression, and one had severe depression (out of 9 patients remaining in the study); at 6 months, no patients suffered from mild or moderate depression, two had moderate severe depression, and one had severe depression (out of 6 patients remaining in the study); finally, at 9 months, one patient had mild depression, one had moderate depression, and one had moderate severe depression, with none having severe depression. Thus, while 100% of the patients who began the study were diagnosed with depression, ranging from mild to severe, only 50% of patients remaining 9 months after completing the methods of invention were found to have depression, none of which were classified as "severe." Thus the methods of the invention successfully reduce both the incidence and severity of depression in patients diagnosed with AUD.
[467] Likewise, as highlighted in FIG. 4, the methods of the invention are effective in reducing mild, moderate, and severe anxiety. Prior to undergoing the methods of the invention, specifically during the screening session, 12 patients were assessed for levels of anxiety. It was found that four patients suffered from moderate anxiety, and eight patients suffered from severe anxiety. Directly after undergoing the detoxification process (noted as "baseline" in FIG. 4), three patients had mild anxiety, two patients had moderate anxiety, and three patients had severe anxiety (out of 11 patients remaining in the study). The patients were then reassessed at 3, 6, and 9 months post-baseline (i.e., after completing a detoxification course, and prior to beginning the MDMA-assisted psychotherapy regimen), wherein it was discovered that, at three months, two patients had mild anxiety, zero patients had moderate anxiety, and 6 patients had severe anxiety (out of the 9 patients remaining in the study); at 6 months, one person had mild anxiety, one person had moderate anxiety, and two people had severe anxiety (out of the 6 patients remaining in the study); at 9 months, one person had mild anxiety, two people had moderate anxiety, and one person had severe anxiety (out of the 6 patients remaining in the study). Thus, while 100% of the patients who began the study were diagnosed with anxiety, ranging from mild to severe, only about 67% of the patients remaining 9 months after completing the methods of the invention were found to have anxiety, only one of which was classified as "severe.- As a result, it can be said that the methods of the invention successfully reduce both the incidence and severity of anxiety in patients diagnosed with AUD.
[468] In some embodiments, the methods of the invention will result in an increase in psychosocial functioning, wherein "psychosocial functioning" refers to one's ability to perform the activities of daily life. In some embodiments, an increase in psychosocial functioning may be measured by obtaining subjective evidence from patients in the form of surveys, questionnaires, and/or other psychosocial functioning assessments, as would be apparent to one of skill. In some such embodiments, the increase in psychosocial functioning is a durable effect, and will be shown at least 1 month, at least 3 months, at least 6 months, at least 9 months, or at least 1 year following treatment. In some embodiments, the patient's psychosocial functioning will improve by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, and about 100%, and values in between. In some most preferred embodiments, the patient's psychosocial functioning will improve by at least 100%, such as at least about 150%, at least about 200%, at least about 250%, at least about 300%, at least about 350%, at least about 400%, at least about 450%, at least about 500%, at least about 550%, at least about 600%, at least about 650%, at least about 700%, at least about 750%, at least about 800%, at least about 850%, at least about 900%, at least about 950%, at least about 1000%, and, in some embodiments, psychosocial functioning will improve will increase by more than about 1000%, as well as values in between.
[469] In some embodiments, the methods of the invention will result in a decrease in use or frequency of a prescription medication. Meaning, in some embodiments, a patient decreases the frequency with which the patient takes a given prescription medication while, in some embodiments, the patient is prescribed a fewer number of medications than the patient was prescribed prior to undergoing the methods of the invention. Additionally, in some embodiments, the patient is both prescribed a fewer number of medications than the patient was prescribed prior to undergoing the methods of the invention, and decreases the frequency with which the patient takes a given prescription medication. As would be apparent to one of skill, in some embodiments, to ascertain whether a patient has decreased prescription medication use, a practitioner may ask the patient to provide information regarding the type, dosage, and frequency of administration of prescription medication.
Alternatively, a practitioner may obtain current prescription records for the patient, or complete a prescription screen through urine or blood testing, as would be apparent to one of skill.
In some such embodiments, the reduction in prescription medication use is a durable effect, and will be shown at least 1 month, at least 3 months, at least 6 months, at least 9 months, or at least 1 year following treatment. In some embodiments, the reduction in prescription medication use (both a decrease in use and frequency of use) persists for about 1 month, about 2 months, about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 12 months, or more than 12 months after completion of the methods of the invention.
[470] In some embodiments, the methods of the invention will result in a decrease in use or frequency of at least one recreational drug. In some embodiments, a recreational drug is prescription drug that is abused, misused, or used illicitly. Meaning, in some embodiments, a patient decreases the frequency with which the patient takes a given recreational drug, while, in some embodiments, the patient takes a fewer number of recreational drugs than the patient took prior to undergoing the methods of the invention. Additionally, in some embodiments, the patient both takes a fewer number of recreational drugs than the patient took prior to undergoing the methods of the invention, and decreases the frequency with which the patient takes a given recreational drug. In some embodiments, as would be apparent to one of skill, a decrease in use or frequency of a recreational drug may be measured by asking the patient to provide information regarding their usage of such substances. Alternatively, a practitioner may conduct a screening for recreational drugs via blood and urine testing, as would be apparent to one of skill. In some such embodiments, the reduction in recreational drug use is a durable effect, and will be shown at least 1 month, at least 3 months, at least 6 months, at least 9 months, or at least 1 year following treatment. In some embodiments, the reduction in recreational drug use (both a decrease in use and frequency of use) persists for about 1 month, about 2 months, about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 12 months, or more than 12 months after completion of the methods of the invention.
[471] In some embodiments, the methods of the invention will result in a reduction in obsessive compulsive thoughts. In some embodiments, the reduction in obsessive compulsive thoughts may be measured by obtaining subjective reports from a patient, wherein the patient discusses the frequency and severity of obsessive compulsive thoughts; by administering to the patient at least one questionnaire or survey, wherein the at least one questionnaire or survey assesses the frequency and/or severity of the patient's obsessive compulsive thoughts; and/or having the patient log the frequency and severity of obsessive compulsive thoughts throughout the duration of the methods of the invention, so a practitioner may objectively determine whether or not the frequency and/or severity of such obsessive compulsive thoughts has reduced. In some such embodiments, the reduction in obsessive compulsive thoughts is a durable effect, and will be shown at least 1 month, at least 3 months, at least 6 months, at least 9 months, or at least 1 year following treatment. In some embodiments, the reduction in obsessive compulsive thoughts will be reduced by between about 5%, to about 100%. In some embodiments, the reduction in obsessive compulsive thoughts will be reduced by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, up to and including 100% (i.e., the patient has substantially no obsessive compulsive thoughts), and values in between
[472] In some embodiments, the methods of the invention will result in a decrease in suicidality. In some embodiments, a decrease in suicidality may be measured by obtaining subjective reports from a patient, wherein the patient discusses the frequency with which they experience suicidal thoughts; by administering to the patient at least one questionnaire and/or survey, wherein the at least one questionnaire and/or survey is capable of quantifying the suicidality of the patient; administering a suicide risk assessment test, suicide ideation risk assessment, or a suiccide severity rating scale, such as the Columbia suicide severity rating scale (C-SSRS); and/or having the patient log the frequency of suicidal thoguhts prior to, during, and after completing the methods of the invention, so that a practitioner may objectibely determine weather or not the patient experienced a reduction in suicidality as a result of the methods of the invention. In some such embodiments, the reduction in suicidality is a durable effect, and will be shown at least 1 month, at least 3 months, at least 6 months, at least 9 months, or at least 1 year following treatment. In some embodiments, suicidality will be reduced by between about 5%, to about 100%. In some embodiments, suicidality will be reduced by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, up to and including 100% (i.e., the patient has substantially no suicidal thoughts), and values in between.
[473] In some embodiments, the methods of the invention will result in an increase in feelings of empathy. In some embodiments, the increase in feelings of empathy may be measured by obtaining subjective reports from a patient, wherein the patient discusses the changes in empathetic feelings and actions they have experienced since completing the methods of the invention; and/or by utilizing questionnaires and/or surveys, wherein the questionnaires and/or surveys assess the subject's empathy through a series of questions. In such embodiments, a practitioner may assess the patient's level of empathy prior to, during, and after undergoing the methods of the invention, to determine whether the patient has experienced an increase in feelings of empathy as a result of the methods of the invention. In some such embodiments, the increase in feelings of empathy is a durable effect, and will be shown at least 1 month, at least 3 months, at least 6 months, at least 9 months, or at least 1 year following treatment. In some embodiments, feelings of empathy will increase by at least about 5%, preferably by at least 25%, more preferably by at least 50%, and most preferably by at least 100%. In some embodiments, feelings of empathy will increase by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, and about 100%, and values in between. In some most preferred embodiments, feelings of empathy will increase by at least 100%, such as at least about 150%, at least about 200%, at least about 250%, at least about 300%, at least about 350%, at least about 400%, at least about 450%, at least about 500%, at least about 550%, at least about 600%, at least about 650%, at least about 700%, at least about 750%, at least about 800%, at least about 850%, at least about 900%, at least about 950%, at least about 1000%, and, in some embodiments, self-compassion will increase by more than about 1000%, as well as values in between.
[474] In some embodiments, the methods of the invention will result in an increase in self-compassion. In some embodiments, the increase in self-compassion may be measured by obtaining subjective reports from a patient, wherein the patient discusses the changes in self-compassion that they have experienced since completing the methods of the invention, and/or by utilizing questionnaires and/or surveys, wherein the questionnaires and/or surveys assess the subject's self-compassion through a series of questions. In such embodiments, a practitioner may assess the patient's self-compassion prior to, during, and after undergoing the methods of the invention, to determine whether the patient has experienced an increase in self-compassion as a result of the methods of the invention. In some such embodiments, the increase in self-compassion is a durable effect, and will be shown at least 1 month, at least 3 months, at least 6 months, at least 9 months, or at least 1 year following treatment. In some embodiments, self-compassion will increase by at least about 5%, preferably by at least 25%, more preferably by at least 50%, and most preferably by at least 100%. In some embodiments, self-compassion will increase by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, and about 100%, and values in between. In some most preferred embodiments, self-compassion will increase by at least 100%, such as at least about 150%, at least about 200%, at least about 250%, at least about 300%, at least about 350%, at least about 400%, at least about 450%, at least about 500%, at least about 550%, at least about 600%, at least about 650%, at least about 700%, at least about 750%, at least about 800%, at least about 850%, at least about 900%, at least about 950%, at least about 1000%, and, in some embodiments, self-compassion will increase by more than about 1000%, as well as values in between.
[475] In some embodiments, the methods of the invention will result in an improvement of at least one symptom of a comorbid psychiatric disorder, wherein the comorbid psychiatric disorder includes, but is not limited to, antisocial personality disorder, borderline personality disorder, depression, anxiety, schizophrenia, attention deficit hyperactivity disorder, bipolar disorder, obsessive compulsive disorder, binge eating disorder, and PTSD. In some embodiments, the improvement of at least one symptom of a comorbid psychiatric disorder may be measured by obtaining subjective reports from a patient, wherein the patient discusses any changes in the at least one symptom of a comorbid psychiatric disorder that they have experienced since completing the methods of the invention; by utilizing questionnaires and/or surveys, wherein the questionnaires and/or surveys assess the severity of the at least one symptom of a comorbid psychiatric disorder through a series of questions;
and/or obtaining objective measurements, as disclosed herein. In some such embodiments, the improvement in at least one symptom of a comorbid psychiatric disorder is a durable effect, and will be shown at least 1 month, at least 3 months, at least 6 months, at least 9 months, or at least 1 year following treatment. In some embodiments, at least one symptom of a comorbid psychiatric disorder will improve by between about 5%, to about 100%. In some embodiments, at least one symptom of a comorbid psychiatric disorder will improve by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100%, and values in between.
Assessments of Quality of Life and Psychosocial Functioning
[476] In some embodiments, quality of life and/or psychosocial functioning are evaluated, monitored, or rated to assess therapeutic outcomes in response to a treatment intervention, such as MDMA-assisted psychotherapy. In some embodiments, quality of life and/or psychosocial functioning can be assessed at one or more points in time prior to, during, after completion of a treatment, such as MDMA-assisted psychotherapy. In some embodiments, quality of life and/or psychosocial functioning can be assessed using subject-reported and/or interviewer-reported questionnaires. Such questionnaires include inquiries related to one or more aspects of a subject's physical, social, and emotional well-being. In some aspects, quality of life and psychosocial functioning assessments can share commonalities, especially when related to a subject's ability to perform the activities of daily living, a subject's engagement in relationships and satisfaction associated with the same, and the interactions of the subject with their workplace and/or community.
[477] In some examples, quality of life and psychosocial functioning can be assessed by questions related to health or improvement of a condition, for example, AUD.
Examples of quality of life and psychosocial functioning questionnaires and their contents are described below for illustrative purposes and should not be construed as limiting. In some cases, the described questionnaires have been used in studies of subjects who display alcohol abuse and dependence (Ugochukwu, 2013).
i. Quality of Life Scale (QoLS)
[478] Although its inquiries are general, e.g., a patient population, the Quality of Life Scale (QoLS) has been used to evaluate quality of life and psychosocial functioning of subjects with various disease states (Burckhardt, 2003). The QOLS may be self-administered or administered by an interviewer. If the interview format is chosen, subjects are provided with a copy of a scale to refer to when deciding on the most appropriate response. Updated versions of the QOLS employ a 7-point scale to rate subject satisfaction. On the 1 to 7 scale, 1 corresponds to "terrible," 2 corresponds to "unhappy," 3 corresponds to "mostly dissatisfied," 4 corresponds to "mixed," 5 corresponds to "mostly satisfied," 6 corresponds to "pleased,"
and 7 corresponds to "delighted" (Burckhardt, 2003).
[479] Subjects are asked to rate various aspects of life, including material and physical well-being, health, relationships with parents, siblings and other relatives, having and raising children, relationship with spouse or significant other, relationships with friends, helping and encouraging others, participating in organizations and public affairs, intellectual development, understanding of self, occupational role, creativity/personal expression, socializing, passive and observational recreation, active and participatory recreation, and independence (doing for yourself). The QOLS scores are added across domains, and a greater total score indicates a higher quality of life.
Health Related Quality of Life (HRQoL)
[480] The Health Related Quality of Life (HRQoL) questionnaire probes the impact of health on a subject's quality of life and psychosocial functioning. Subjects respond with graded, i.e.
on a point scale, or non-graded responses to questions in three domains:
healthy days, activity limitations, and healthy day symptoms (CDC, 2016). Examples of non-graded responses include providing the number of days physical health was not good, the number of days mental health was not good, and the number of days when health interfered with daily activities.
Questions related to activity limitations evaluate the ability to fulfill personal care needs and the demands of a subject's routine. Healthy day symptoms include inquiries into pain, depression, anxiety, rest and sleep, and a general assessment of health and energy level (Stanford Sparqtools: Health-Related Quality of Life Scale).
iii. World Health Organization Quality of Life
[481] World Health Organization Quality of Life instruments (WHOQOL) include, for example, the WHOQOL-100 and its abbreviated version, WHOQOL-BREF (26 items).
The WHOQOL was developed by fifteen international field centres to develop a ross-culturally applicable quality of life assessment. The surveys may be self-administered or interviewer-administered. The WHOQOL-BREF prompts scoring of four domains related to quality of life:
physical health, psychological, social relationships, and environment. It also includes overall quality of life and general health. Though abbreviated, results of the WHOQOL-BREF
correlate highly to WHOQOL-100 domain scores, as calculated on a four domain structure (Development of the World Health Organization WHOQOL-BREF quality of life assessment, 1998).
[482] In brief, scoring of WHOQOL-BREF entails examining two items separately (an individual's overall perception of quality of life and an individual's overall perception of their health) in addition to examining the four domains. The four domain scores denote an individual's perception of quality of life in each particular domain. Higher domain scores denote higher quality of life. The average score of items within each domain is used to calculate the domain score. If it is desired to compare domain results to those of the WHOQOL-100, the mean scores are then multiplied by 4 (WH0,1996).
iv. Munich Quality-of-life Dimension List (MLDL)
[483] The Munich Quality-of-life Dimension List (MLDL) allows a subject to self-assess the level of personal satisfaction in all basic areas of life. The MLDL includes 19 different items relevant to quality of life, including mental, physical, social and everyday-life dimensions during the last week on one overall scale and four subscales using Liken scales ranging from 0 (very unsatisfied/very unimportant) to 10 (very satisfied/very important).
[484] Exemplary quality of life domains that may be assessed include "overall," "physical aspects," "psychological aspects," "social life," and "everyday life." In addition to rating one's satisfaction, the scale also prompts the respondent to evaluate the personal relevance of every item using Likert scales from 0 (very unimportant) to 10 (very important). A
calculation algorithm may be used to determine relevance-weighted satisfaction scores. In the absence of differences in relevance ratings in the group comparisons of interest, unweighted satisfaction scores have been used (Frischknecht, 2013).
v. Medical Outcomes Study Short Form (SF-36)
[485] The Medical Outcomes Study Short Form (SF-36) is an example of a questionnaire that can be used as a self-reported and an observer-reported survey. The survey was designed for self-administration by persons 14 years of age and older, and for administration by a trained interviewer in person or by telephone. The SF-36 evaluates eight health concepts on one multi-item scale: 1) limitations in physical activities because of health problems;
2) limitations in social activities because of physical or emotional problems; 3) limitations in usual role activities because of physical health problems; 4) bodily pain; 5) general mental health, e.g., psychological distress and well-being; 6) limitations in usual role activities because of emotional problems; 7) vitality, e.g., energy and fatigue; and 8) general health perceptions (Ware, 1992). Subsequent versions of the SF-36 have been developed that vary in length, e.g., the SF-6D and the SF-12, and rating scale, for example, reducing six-level response scales to a five-level scale.
[486] A high or relatively higher SF-36 score indicates a better health state.
After the survey has been completed, items and scales are scored in three steps: 1) item recoding, for the 10 items that require recoding; 2) computing scale scores by summing across items in the same scale (raw scale scores); and 3) transforming raw scale scores to a 0-100 scale (transformed scale scores). It is recommended that recoding and scale scoring be performed by a computer using the appropriate algorithms (Ware, 1993).
vi. Quality of Life Enjoyment and Satisfaction Questionnaire (Q-LES-Q)
[487] The Quality of Life Enjoyment and Satisfaction Questionnaire (Q-LES-Q) prompts the respondent to rate satisfaction with various aspects of life in the past week, including mood, health, relationships, and economic status, among others. A numeric scale is correlated with qualitative measurements, for example, 1=Very Poor to 5=Very Good. Higher scores indicate greater satisfaction with different aspects of life.
vii. EuroQoL-5D (EQ-5D)
[488] The EuroQoL-5D (EQ-5D) assesses five domains: mobility, usual activities, self-care, pain or discomfort, and anxiety or depression. Responses in each domain are rated on a scale from 1 to 3, where 1 indicates no problems, 2 indicates moderate problems, and 3 indicates extreme problems. The ratings in each domain constitute the five-digit number collectively known as the EQ-5D self-reported health state (Gunther, 2007).
viii. Life Situation Survey (LSS)
[489] The Life Situation Survey (LSS) is a generic scale that assesses an individual's current functioning using 20 QoL indicators, including work, leisure, nutrition, sleep, social nurturance, earnings, health, love/affection, environment, self-esteem, security, public support, stress, mobility, autonomy, energy level, social support, mood/affect, outlook, and egalitarianism. Variables are rated on a scale from 1 to 7, with total scores ranging from 20 to 140. Scores of less than 90 correspond to a poor quality of life, which can be defined as greater psychosocial impairment (Foster, 2000).
ix. Nottingham Health Profile (NHP)
[490] The Nottingham Health Profile (NHP) is a self-evaluated questionnaire that measures a subject's perceived physical health and emotional and social problems. This scale evaluates the domains of energy, physical functioning/mobility, pain, sleep, affective control, and social isolation. Each section is rated with a score from 0 to 100, where 100 represents the most severe psychosocial/health-related dysfunction (Hunt, 1980).
x. Clinical Global Impression ¨ Improvement Scale (CGI-I)
[491] The Clinical Global Impression ¨ Improvement Scale (CGI-I) facilitates an interviewer's or an observer's, e.g., a clinician's, rating of a subject's global functioning prior to and after initiating a treatment intervention. This facilitates tracking of clinical progress over time. All available information, including a knowledge of the patient's history, psychosocial circumstances, symptoms, behavior, and the impact of the symptoms on the subject's ability to function can be considered when rating using the CGI-I. The CGI-I comprises two companion one-item measures that evaluate the severity of psychopathology on a scale from 1 to 7 and changes from the initiation of treatment on a similar seven-point scale (Busner, 2007).
xi. Sleep Quality Scale (SQS)
[492] The Sleep Quality Scale (SQS) consists of 28 items to self-assess six domains of sleep quality, including daytime symptoms, restoration after sleep, problems initiating and maintaining sleep, difficulty waking, and sleep satisfaction. Respondents use a four-point, Likert-type scale to indicate the frequency of certain sleep behaviors, i.e., 0 = "few," 1 =
"sometimes," 2 = "often," and 3 = "almost always". Scores on items belonging to factors 2 and (restoration after sleep and satisfaction with sleep) are reversed before being tallied. Total scores can range from 0 to 84, with higher scores indicative of acute sleep problems (Yi, 2006).
xii. Pittsburgh Sleep Quality Index (PSQI)
[493] The Pittsburgh Sleep Quality Index (PSQI) is a self-rated questionnaire, which is used to assess sleep quality, including sleep disturbances, over a 1-month time interval. Nineteen individual items generate seven "component" scores in the areas of subjective sleep quality, sleep latency, sleep duration, habitual sleep efficiency, sleep disturbances, use of sleeping medication, and daytime dysfunction. The scores across the seven components are added to yield one global score (Buysse, 1989).
xiii. Leeds Sleep Evaluation Questionnaire (LSEQ)
[494] The Leeds Sleep Evaluation Questionnaire (LSEQ) uses ten 100 mm visual analogue scale items in four areas related to the ease of getting to sleep (GTS), the perceived quality of sleep (QOS), the ease of awakening from sleep (AFS), and the integrity of early-morning behavior following wakefulness (BFW). A subject is instructed to answer each question by placing a vertical mark on a line, i.e., the "scale." If no change was experienced, a subject would place a mark in the middle of the line. If a change was experienced, then the position of the subject's mark indicates the nature and extent of the change, for example, large charges are represented as near the ends of the line and small changes near the middle of the line (Parrott, 1980) Assessments of Alcohol and Drug Use and Cravings
[495] In some embodiments, use and/or cravings of alcohol and/or drugs are evaluated, monitored, or rated to assess therapeutic outcomes in response to a treatment intervention, for example, MDMA-assisted psychotherapy. In some embodiments, aspects of alcohol and drug use and cravings can be assessed at one or more points in time prior to, during, after completion of a treatment, such as MDMA-assisted psychotherapy. In some embodiments, alcohol and drug use and cravings are assessed using subject-reported and/or interviewer-reported questionnaires. Alcohol- and drug-related inquiries include, but are not limited to, evaluations of alcohol use, alcohol cravings, use of prescribed medication(s), number and frequency of use of recreational drugs, cravings relating to MDMA, and use of illicit ecstasy following MDMA-assisted psychotherapy. Examples of instruments for monitoring or rating alcohol and drug use and cravings and their contents are described below for illustrative purposes and should not be construed as limiting.
i. Alcohol Use and Cravings
[496] In some embodiments, alcohol acquisition and consumption can be evaluated using a variety of mediums, including interviewing or asking a subject about drinking behavior, administering a self-assessment on drinking behavior, e.g., a questionnaire, or detecting a level of alcohol, e.g., blood alcohol content, such as from a breath, body fluid, or tissue specimen.
In some embodiments, cravings for alcohol include, for example, experiencing anticipation and/or desire for consuming alcohol. In some embodiments, alcohol cravings manifest as alcohol-seeking behavior. In some embodiments, cravings may be prompted or triggered by a variety of stimuli including, for example, alcohol withdrawal, the sight or smell of alcohol, interactions with people who are drinking, or even reliving a memory where drinking was involved.
[497] In some non-limiting examples, alcohol use and/or alcohol cravings can be evaluated using the Severity of Alcohol Dependence Questionnaire (SADQ) (Stockwell, 1983), The Alcohol Dependence Syndrome section of the SCID-5-CT (Clinical Trials Version) (First, 2015), The Alcohol Timeline Follow Back (TLFB) (Sobell, 2001), The Clinical Institute Withdrawal Assessment for Alcohol¨Revised Version (CIWA¨Ar) (Saitz, 1994), The Penn Alcohol Craving Scale (PACS) (Flannery, 1999), and the Obsessive Compulsive Drinking Scale (OCDS) (Anton, 1995).
Drug Use and Cravings
[498] In some embodiments, drug use, for example use of prescription or recreational drugs, can be evaluated using a variety of mediums, including interviewing or asking subject's prescribing physician about prescribed medications, interviewing or asking a subject about their drug use, administering a self-assessment on drug use, e.g., a questionnaire, or detecting a level of a drug and/or its metabolite(s) using a variety of methods known to one of skill in the art from, for example, a breath, body fluid, or tissue specimen
[499] In some embodiments, prescription drug use can be evaluated using a variety of mediums, including interviewing or asking the subject's prescribing physician about prescribed medications and/or interviewing or asking the subject about the same. In some embodiments, use of drugs, such as prescription drugs, can be evaluated using measures of adherence and/or compliance. In some embodiments, measures of adherence and/or compliance include, for example, use of electronic medication packaging (EMP) devices, pill counting, clinician assessments, and self-report (Lam, 2015). In some embodiments, use of drugs, such as prescription drugs, can be evaluated using self-reported or interviewer-reported questionnaires.
For example, the Medication Adherence Questionnaire (MAQ) (Morisky, 1986) has been used to identify and predict patterns of adherence (Culig, 2014).
[500] In some embodiments, use of drugs, such as recreational drugs, can be evaluated using self-reported or interviewer-reported questionnaires. For example, the Drug Abuse Screening Test (DAST-10 and DAST-20) is a self-reported 20-item questionnaire that assesses the extent of the problems related to drug misuse, using yes or no responses. The DAST
total score is computed by summing all items (Villalobos-Gallegos, 2016).
[501] In some embodiments, cravings for drugs, such as recreational drugs, include, for example, experiencing anticipation and/or desire for consuming such substances. In some embodiments, cravings for drugs, such as recreational drugs, manifest as drug-seeking behavior. In some embodiments, drug cravings are determined using self-reported questionnaires. In some embodiments, drug cravings can be identified or measured using reinforcement -proxies," drug self-administration, psychophysiological responding, neurobiological responding, cognitive processing, and expressive behavior (Sayette, 2000).
[502] In some embodiments, use and/or cravings for MDMA or ecstasy are determined using one or more of the methods described herein. In some embodiments, use and/or cravings for MDMA or ecstasy are determined by asking a subject, such as a subject who will, is, or has participated in an MDMA-assisted psychotherapy treatment regimen, about their use of MDMA and cravings for the same. In some embodiments, use and/or cravings for MDMA or ecstasy are determined using a self-reported assessment. In some embodiments, use and/or cravings for MDMA or ecstasy are determined using other methods, for example, reinforcement "proxies," drug self-administration, psychophysiological responding, neurobiological responding, cognitive processing, and expressive behavior (Sayette, 2000).
[503] These and other benefits, advantages, and improvements of the invention will be known by reference to the following claims.

REFERENCES
1. Anton RF, Moak DH, Latham P. (1995) The Obsessive Compulsive Drinking Scale: a self-rated instrument for the quantification of thoughts about alcohol and drinking behavior.
Alcohol Clin Exp Res. 1995 Feb;19(1):92-9.
2. Anton, R. F., O'Malley, S. S., Ciraulo, D. A., Cisler, R. A., Couper, D., Donovan, D. M., et al. for the COMBINE Study Research Group (2006). Combined pharmacotherapies and behavioural Interventions for alcohol dependence. The COMBINE study: a randomized controlled trial. JAMA, 293, 2003-2017.
3. APA 2013. Diagnostic and statistical manual of mental disorders: DSM-5.
Washington D.C.: American Psychiatric Association.
4. Arza et al. Formulation and evaluation of swellable and floating gastroretentive ciprofloxacin hydrochloride tablets, AAPS PharmSciTech., 10(1):220-226 (2009).
5. Back-Madruga C, Boone KB, Chang L et al. (2003) Neuropsychological effects of 3,4-methylenedioxymethamphetamine (MDMA or ecstasy) in recreational users. Clin Neuropsychol 2003; 17: 446 - 59.
6. Barrett, F. S., Johnson, M. W., & Ciriffiths, R. R. (2015). Validation of the revised Mystical Experience Questionnaire in experimental sessions with psilocybin. Journal of psychopharmacology (Oxford, England), 29(11), 1182-1190 7. Berge et al. (1977) "Pharmaceutical Salts," J. Pharm. Sci. 66:1-19.
8. Bhattachary S, Powell JH (2001) Recreational use of 3,4-methylenedioxymethamphetamine (MDMA) or "ecstasy": evidence for cognitive impairment. Psychol Med 31: 647-658.
9. Bierer, L.M. et al., Intergenerational Effects of Maternal Holocaust Exposure on FKBP5 Methylation, American Journal of Psychiatry 2020 177:8, 744-753.
10. Bishehsari, F., Magno, E., Swanson, G., Desai, V., Voigt, R. M., Forsyth, C. B., &
Keshavarzian, A. (2017). Alcohol and Gut-Derived Inflammation. Alcohol research:
current reviews, 38(2), 163-171.
11. Bogenschutz MP, Forcehimes AA, Pommy JA, Wilcox CE, Barbosa PC, Strassman RJ.
(2015) Psilocybin-assisted treatment for alcohol dependence: a proof-of-concept study. J
Psychopharmacol. 2015 Mar;29(3):289-99.
12. Bolton D (2008). What is Mental Disorder?: An Essay in Philosophy, Science, and Values.
OUP Oxford. p. 6. ISBN 978-0-19-856592-5.
13. Bowen, S., Chawla, N., Collins, S. E., Witkiewitz, K., Hsu, S., Grow, J., Clifasefi, S., Garner, M., Douglass, A., Larimer, M. E., & Marlatt, A. (2009). Mindfulness-based relapse prevention for substance use disorders: a pilot efficacy trial. Substance abuse, 30(4), 295-305. https://doi.org/10.1080/08897070903250084.
14. Brunner D, Hen R. (1997) Insights into the neuro- biology of impulsive behavior from serotonin receptor knockout mice. Ann NY Acad Sci 1997;836:81-105.
15. Burckhardt, C. S., & Anderson, K. L. (2003). The Quality of Life Scale (QOLS): reliability, validity, and utilization. Health and quality of life outcomes, 1, 60 16. Busner, J., & Targum, S. D. (2007). The clinical global impressions scale:
applying a research tool in clinical practice. Psychiatry (Edgmont (Pa. : Township)), 4(7), 28-37.
17. Buysse DJ, Reynolds CF 3rd, Monk TH, Berman SR, Kupfer DJ. (1989) The Pittsburgh Sleep Quality Index: a new instrument for psychiatric practice and research.
Psychiatry Res.
1989 May;28(2):193-213.
18. Carhart-Harris RL et at (2013) The effect of MDMA on recollecting autobiographical memories: an fMRI study with implications for 1VIDMA-assisted psychotherapy.
Int J
Neuropsychoph arm a col ogy: Online: http://www.ncbi .nlni.nih.gov/pubmed/2434539S
19. Carhart-Harris RL, Murphy K, Leech R, Erritzoe D, Wall MB, Ferguson B, Williams LTJ, Roseman L, Brugger S, De Meer I, Tanner M, Tyacke R, Wolff K, Sethi A, Bloomfield MAP, Williams TM, Bolstridge M, Stewart L, Morgan C, Newbould RD, Feilding A, Curran BY, Nutt DJ et al. (2015) The Effects of Acutely Administered 3,4-Methylenedioxymethamphetamine on Spontaneous Brain Function in Healthy Volunteers Measured with Arterial Spin Labeling and Blood Oxygen Level-Dependent Resting State Functional Connectivity, BIOLOGICAL PSYCHIATRY, Vol: 78, Pages: 554-562 20. Carhart-Harris RL, Wall MB, Erritzoe D, Kaelen M, Ferguson B, De Meer I, Tanner M3 Bloomfield M, Williams TM, Bolstridge M, Stewart L, Morgan C, Newbould RD, Feilding A, Curran BY, Nutt DJ (2013) The effect of acutely administered MDMA on subjective and BOLD fMRI responses to favourite and worst autobiographical memories. Int J of Neuropsychopharmacology 17: 527-40 21. Cargill Z, Kattiparambil 5, Hansi N, et al (2020) Severe alcohol-related liver disease admissions post-COVID-19 lockdown: canary in the coal mine? Frontline Gastroenterology Published Online First: 26 October 2020. doi: 10.1136/flgastro-2020-101693.
22. Carvalho M, Pontes H, Remido F, Bastos ML, Carvalho F. Mechanisms underlying the hepatotoxic effects of ecstasy. Curr Pharm Biotechnol. 2010 Aug;11(5):476-95.
doi:
10.2174/138920110791591535. PMID: 20420570.
23. Castillo-Carniglia, Alvaro et al., Psychiatric comorbidities in alcohol use disorder, The Lancet Psychiatry, Vol. 6, Issue 12, 1068-1080.
24. Center for Drug Evaluation and Research. (2013, September 25). Drug interactions: What you should know. U.S. Food and Drug Administration. Retrieved December 23, 2021, from https://www.fda.gov/drugs/resources-you-drugs/drug-interactions-what-you-should-know 25. Centers for Disease Control and Prevention. (2016). Health-related quality of life (HROOL). Retrieved from: https://www.cdc.gov/hrqol/methods.htm.
26. Centers for Disease Control and Prevention. (2019, December 30). Binge drinking is a serious but preventable problem of excessive alcohol use. Centers for Disease Control and Prevention. Retrieved Dec. 23, 2021, www.cdc.gov/alcohol/fact-sheets/binge-drinking.htm 27. Chabrol H. & Oehen, P. (2013) MDMA assisted psychotherapy found to have a large effect for chronic post-traumatic stress disorder. J Psychopharmacol; s27(9):865-6.
28. Clay, J.M., Parker, M.O. (2020) Alcohol use and misuse during the COVID-19 pandemic: a potential public health crisis? The Lancet: Public Health, Correspondence, Volume 5, ISSUE 5, e259, May 01, 2020.

29. What is tapering? Coalition Recovery. (2019, October 2). Retrieved December 23, 2021, from https://www.coalitionrecovery.com/rehab-blog/what-is-tapering/
30. Cozzi NV, Sievert MK, Shulgin AT, et al. (1999) Inhibition of plasma membrane monoamine transporters by beta-ketoamphetamines. Eur J Pharmacol 1999;381:63-9.
31. Culig J, Leppee M. From Morisky to Hill-bone; self-reports scales for measuring adherence to medication. Coll Antropol. 2014 Mar;38(1):55-62. PMID: 24851597.
32. Curran L, Andrykowski MA, Studts, JL. (1995). Short Form of the Profile of Mood States (POMS-SF): Psychometric information. Psychological Assessment. 7 (1): 80-83 33. Currie C, Davies A, Blunt I, Anti C, Bardsley M (2015) Alcohol-specific activity in hospitals in England. Research report. The Nuffield Trust.
34. Curry, D. W., Young, M. B., Tran, A. N., Daoud, G. E., & Howell, L. L.
(2018). Separating the agony from ecstasy: R(-)-3,4-MDMA has prosocial and therapeutic-like effects without signs of neurotoxicity in mice. Neuropharmacology, 128, 196-206.
https://doi. org/10.1016/j .ne uropharm.2017. 10.003.
35. Davis, AK., Barrett, F.S., Griffiths, R.R. (2020). Psychological flexibility mediates the relations between acute psychedelic effects and subjective decreases in depression and anxiety. Journal of Contextual Behavioral Science. 15, 39-45.
36. Davis, M.H. (1980). A multidimensional approach to individual differences in empathy.
JSAS Catalog of Selected Documents in Psychology, 10, 85.
37. de la Torre R, Farre M, Roset PN, Pizarro N, Abanades S, Segura M, Segura J, Cami J.
Human pharmacology of MDMA: pharmacokinetics, metabolism, and disposition.
Ther Drug Monit. 2004 Apr;26(2):137-44. doi: 10.1097/00007691-200404000-00009.
PMID:
15228154.
38. Development of the World Health Organization WHOQOL-BREF quality of life assessment. The WHOQOL Group. Psycho' Med. 1998 May,28(3):551-8. doi:
10.1017/s0033291798006667. P1V11D: 9626712.
39. Ding, W. X., & Yang, L. (2019). Alcohol and drug-induced liver injury:
Metabolism, mechanisms, pathogenesis and potential therapies*. Liver research, 3(3-4), 129-131.
https://doi.org/10.1016/j.livres.2019.11.006 40. Ditman, KS., Bailey, J.J., 1967. Evaluating LSD as a psychotherapeutic agent. In:
Abramson, H. (Ed.), The Use of LSD in Psychotherapy and Alcoholism.
BobbsMerrill, New York, NY, pp. 74e80.
41. Dittrich A. (1998). The standardized psychometric assessment of altered states of consciousness (ASCs) in humans. Pharmacopsychiatry. 31, Suppl 2:80-4. doi:
10.1055/s-2007-979351. PMID: 9754838.
42. Doblin R1, Greer G, Holland J, Jerome L, Mithoefer MC, Sessa B. (2014) A
reconsideration and response to Parrott AC (2013) "Human psychobiology of MDMA
or 'Ecstasy': an overview of 25 years of empirical research". Hum Psychopharmacol. 2014 Mar;29(2):105-8.
43. Egerer G, Stickel F, Seitz HK, 45 - Alcohol and the Gastrointestinal Tract, Comprehensive Handbook of Alcohol Related Pathology, Academic Press, 2005, Pages 557-570, ISBN

9780125643702, https://doi.org/10.1016/B978-012564370-2/50047-7.
(https://www.sciencedirect.com/science/article/pii/B9780125643702500477) 44. Evren, C., Durkaya, M., Dalbudak, E., celik, S., cetin, R., & cakmak, D.
(2010). Factors related with relapse in male alcohol dependents: 12 months follow-up study.
Dasiinen Adam: The Journal of Psychiatry and Neurological Sciences,23(2), 92-99.
45. First MB, et al. (1997) Structured Clinical Interview for DSM-IV
Personality Disorders, (SCID-II) Washington, DC: American Psychiatric Press; 1997.
46. First MB, Williams JBW, Karg RS, Spitzer RL. (2015) Structured Clinical Interview for DSM-5 Disorders, Clinical Trials Version (SCID-5-CT). Arlington, VA, American Psychiatric Association, 2015 47. Flannery BA, Volpicelli JR and Pettinati HM (1999) Psychometric properties of the Penn Alcohol Craving Scale. Alcohol Clin Exp Res 23: 1289-1295.
48. Foster JH, Peters T.1, Marshall EJ. Quality of life measures and outcome in alcohol-dependent men and women. Alcohol. 2000 Aug;22(1):45-52. doi: 10.1016/s0741-8329(00)00102-6. PMID: 11109027.
49. Frischknecht U, Sabo T, Mann K. (2013) Improved Drinking Behaviour Improves Quality of Life: A Follow-Up in Alcohol-Dependent Subjects 7 Years After Treatment, Alcohol and Alcoholism, 48(5):579-584.
50. Gamma A, Buck A, Berthold T, Liechti ME, Vollenweider FX. (2000) 3,4-Methylenedioxymethamphetamine (MDMA) modulates cortical and limbic brain activity as measured by [H(2)(15)01-PET in healthy humans. Neuropsychopharmacology 23: 388-95.
51. Gouzoulis-Mayfrank E., Thimm B., Rezk M., Hensen G., Daumann J. (2003).
Memory impairment suggests hippocampal dysfunction in abstinent ecstasy users. Prog.
Neuropsychopharmacol. Biol. Psychiatry 27, 819-827 52. Green (1996) The Trauma History Questionnaire (THQ) 53. Greer GR, Tolbert R. (1986) Subjective Reports of the Effects of MDMA
in a Clinical Setting. Journal of Psychoactive Drugs. Vol. 18(4) Oct-Dec, 1986 54. Greer GR, Tolbert R. (1998) A method of conducting therapeutic sessions with MDMA.
30: 371-379 Journal of Psychoactive Drugs.
55. Grof (2008) LSD Psychotherapy (Ben Lomond, CA: Multidisciplinary Association for Psychedelic Studies).
56. Gunther 0, Roick C, Angermeyer MC, Konig HR. The EQ-5D in alcohol dependent patients: relationships among health-related quality of life, psychopathology and social functioning. Drug Alcohol Depend. 2007 Jan 12;86(2-3):253-64. doi:
10.1016/j.drugalcdep.2006.07.001. Epub 2006 Aug 22. PMID: 16930855.
57. Hallgren KA, Witkiewitz K. Missing data in alcohol clinical trials: a comparison of methods. Alcohol Clin Exp Res. 2013 Dec;37(12):2152-60.
58. Halpern JH, Sherwood AR, Hudson J1, Gruber S, Kozin D, Pope HG, Jr.
(2004) Residual neurocognitive features of long-term ecstasy users with minimal exposure to other drugs.
Drug and Alcohol Dependence. 75, 135-147 59. Hamer, R. and Simpson, P., 2009. Last Observation Carried Forward Versus Mixed Models in the Analysis of Psychiatric Clinical Trials. American J. Psychiatry, 166(6):639-41 60. Hanson, K. L., & Luciana, M. (2010). Neurocognitive impairments in MDMA
and other drug users: MDMA alone may not be a cognitive risk factor. Journal of Clinical and Experimental Neuropsychology, 32, 337-349.
61. Hanson, K.L., & Luciana, M. (2004). Neurocognitive function in users of MDMA: the importance of clinically significant patterns of use. Psychological Medi., 34, 62. Harris DS, Baggott M, Mendelson JH, Mendelson JE, Jones RT. (2002) Subjective and hormonal effects of 3,4- methylenedioxymethamphetamine (MDMA) in humans.
Psychopharmacology (Berl); 162(4): 396-405.
63. Harris, R. (2009). ACT Made Simple: An Easy-lb-Read Primer on Acceptance and Commitment Therapy. Oakland, CA: New Harbinger.
64. Hendershot, C. S., Witkiewitz, K., George, W. H., & Marlatt, G. A. (2011).
Relapse prevention for addictive behaviors. Substance abuse treatment, prevention, and policy, 6, 17. https://doi.org/10.1186/1747-597X-6-17.
65. Holland, Julie; Metzner, Ralph; Adamson, Sophia (2001). Ecstasy: the complete guide; a comprehensive look at the risks and benefits of MDMA. Rochester, Vt., Park Street Press.
ISBN 978-0-89281-857-0.
66. Holze, F., Vizeli, P., Muller, F., Ley, L., Duerig, R., Varghese, N., Eckert, A., Borgwardt, S., & Liechti, M. E. (2020). Distinct acute effects of LSD, MDMA, and D-amphetamine in healthy subjects. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology, 45(3), 462-471. https://doi.org/10.1038/s41386-019-67. FIM Government (2012) The Government's Alcohol Strategy. Secretary of State for the Home Department HM Government. Page 3.
68. HSCIC (Health and Social Care Information Centre) (2014) Statistics on Alcohol -England, 2014. http://www.hscic.gov.uk/catalogue/PUB14184 69. Hsu SH, Grow J, Marlatt GA. (2008) Mindfulness and addiction. In:
Galanter M, Kaskutas LA, editors. Recent Developments in Alcoholism. Vol. 18. pp. 229-250.
70. Hunt SM, McKenna SP, McEwen J, et alA quantitative approach to perceived health status:
a validation study.Journal of Epidemiology & Community Health 1980;34:281-286.
71. Husain, K., Ansari, R. A., & Ferder, L. (2014). Alcohol-induced hypertension: Mechanism and prevention. World journal of cardiology, 6(5), 245-252.
https://doi .org/10.4330/wj c.v6. i 5.245 72. Hysek CM, Domes G, Liechti ME. (2012a) MDMA enhances "mind reading" of positive emotions and impairs "mind reading" of negative emotions. 222: 293-302.Psychopharmacology.
73. Hysek CM, et al. (2012b) Duloxetine inhibits effects of MDMA
("ecstasy") in vitro and in humans in a randomized placebo-controlled laboratory study. PLoS One. ;7(5):
74. Hysek, C.M., et al. (2013) MDMA enhances emotional empathy and prosocial behavior.
Soc Cogn Affect Neurosci. Nov;9(11):1645-52.

75. Jaiswal et al., Nanoemulsion: an advanced mode of drug delivery system, Biotech 3(5):123-27 (2015).
76. Jerome, L, Schuster, S & Berra Yazar-Klosinski, B. (2013) Can MDMA Play a Role in the Treatment of Substance Abuse? Current Drug Abuse Reviews, 2013, 6, 000-000 77. Johnson, Richards, & Griffiths (2008). Human hallucinogen research:
guidelines for safety.
Journal of psychopharmacology, 22(6), 603-620.
78. Johnson MW, Garcia-Romeu A, Cosimano MP, Griffiths RR. (2014) Pilot study of the 5-HT2AR agonist psiloeybin in the treatment of tobacco addiction.
79. Kabat-Zinn, J., Massion, A. 0., Kristeller, J., Peterson, L. G., Fletcher, K. E., Pbert, L., Lenderking, W. R., & Santorelli, S. F. (1992). Effectiveness of a meditation-based stress reduction program in the treatment of anxiety disorders. The American journal of psychiatry, 149(7), 936-943. https ://doi org/10.1176/ajp. 149. 7.936.
80. Kabat-Zinn J. Wherever You Go, There You Are: Mindfulness Meditation in Everyday Life.
New York: Hyperion; 1994.
81. Kaelen, M. et al. (2015) LSD enhances the emotional response to music.
Psychopharmacology: 232 (19) 82. Kiluk, B. D., Dreifuss, J. A., Weiss, R. D., Morgenstern, J., &
Carroll, K. M. (2013). The Short Inventory of Problems - Revised (SIP-R): Psychometric properties within a large, diverse sample of substance use disorder treatment seekers. Psychology of Addictive Behaviors: Journal of the Society of Psychologists in Addictive Behaviors, 27(1), 307-314.
83. Krampe, H., Stawicki, S., Wagner, T., Bartels, C., Aust, C., Ruther, E., Poser, W., Ehrenreich, H. (2006). "Follow-up of 180 Alcoholic Patients for up to 7 Years After Outpatient Treatment: Impact of Alcohol Deterrents on Outcome". Alcoholism:
Clinical and Experimental Research 30(1): 86-95.
84. Kranzler HR, Soyka M. Diagnosis and Pharmacotherapy of Alcohol Use Disorder: A
Review. JAMA. 2018;320(8):815-824. doi:10.1001/jama.2018.11406 85. Kroenke K, Spitzer RL, Williams JB. (2001) The PHQ-9: validity of a brief depression severity measure. J Gen Intern Med. Sep;16(9):606-13.
86. Kouimtsidis C (2017) Structured Preparation before Alcohol Detoxification; A Shift from the Current Treatment Paradigm. J Addict Res Ther 8:326. doi:10.4172/2155-6105.1000326 87. Krupitsky, E.M. & Grinenko, A.Y. (1997) Ketamine psychedelic therapy (KPT): A review of the results of ten years of research. J. Psychoactive Drugs 29 (2), 165 -88. Kurland, A.A., Unger, S., Shaffer, J.W., Savage, C., 1967. Psychedelic therapy utilizing LSD in the treatment of the alcoholic patient: a preliminary report. Am. J.
Psychiatry 123 (10), 1202e1209.
89. Kurland, A.A., Savage, C., Pahnke, W.N., Grof, S., Olsson, J.E., 1971.
LSD in the treatment of alcoholism. In: Vinar, 0., Votava, Z., Bradley, P.B. (Eds.), Advances in Neuropsychopharmacology: Proceedings of the 7th Congress of the Collegium International Neuropsychopharma Cologicum. North-Holland, Amsterdam, pp.
361e372.

90. Kuypers KP, Ramaekers JG. (2007) Acute dose of MDMA (75 mg) impairs spatial memory for location but leaves contextual processing of visuospatial information unaffected.
Psychopharmacology (Berl) 2007; 189(4): 557-63.
91. Lachman et al., Theory and Practice of Industrial Pharmacy (1986).
92. Lam, W. Y., & Fresco, P. (2015). Medication Adherence Measures: An Overview. BioMed research international, 2015, 217047. https://doi.org/10.1155/2015/217047 93. Lavelle A, Honner V, Docherty JR. (1999) Investigation of the prejunctional a1pha2-adrenoceptor medi- ated actions of MDMA in rat atrium and vas deferens. Br J
Pharmacol 1999;128:975-80.
94. Liberman et al. Pharmaceutical Dosage Forms, 2 Ed., Vol. 1, pp. 209-214 (1990).
95. Liechti ME, Gamma A, Vollenweider FX. (2001) Gender differences in the subjective effects of MDMA. Psychopharmacology (Berl) 2001; 154(2): 161-8.
96. Lingford-hughes, A.R., Welch, S., Peters, L. and JNutt, D.j. (2012) BAP
updated guidelines: evidence-based guidelines for the pharmacological management of substance abuse, harmful use, addiction and comorbidity: recommendations from bap. J
psychopharmacol. 2012 jul;26(7):899-952.
97. Lowry, R. (2001-2013). Significance of the difference between two correlation coefficients. Retrieved from http://vassarstats.net/rdiff.html 98. Malcolm R, Roberts JS, Wang W, Myrick H, Anton RF. Multiple previous detoxifications are associated with less responsive treatment and heavier drinking during an index outpatient detoxification. Alcohol. 2000 Nov;22(3):159-64. PubMed PlVIID:
11163123.
99. Marcus, M. T., & Zgierska, A. (2009). Mindfulness-Based Therapies for Substance Use Disorders: Part 1 (Editorial). Substance Abuse: Official Publication of the Association for Medical Education and Research in Substance Abuse, 30(4), 263.
100. Marlatt, G.A & Gordon, J.R. (1985). Relapse prevention: Maintenance strategies in the treatment of addictive behaviors. New York: Guilford Press.
101. Mas M, Farre M, de la Torre R, et al. (1999) Cardiovascular and neuroendocrine effects and pharmacokinetics of 3, 4- methylenedioxymethamphetamine in humans. J Pharmacol Exp Ther; 290(1): 136-45.
102. Mayo-Smith MF. (1997) Pharmacological management of alcohol withdrawal. A
meta-analysis and evidence-based practice guideline. American Society of Addiction Medicine Working Group on Pharmacological Management of Alcohol Withdrawal. JAMA. 1997 Jul 9;278(2):144-51. PubMed PMID 9214531.
103. McCulloch, D. E., Grzywacz, M. Z., Madsen, M. K., Jensen, P. S., Ozenne, B., Armand, S., Stenbxk, D. S. (2021, December 14). A Quantitative and Qualitative Report of Psilocybin Induced Mystical-Type Experiences and Their Relation to Lasting Positive Effects. h&lps://doi.org/10.3 I 23 4losf ioixqz112 104. McLean, JR., MacDonald, D.C., Byrne, U.P., Hubbard, A.M., 1961. The use of LSD-25 in 105. the treatment of alcoholism and other psychiatric problems. Q. J. Stud.
Alcohol 22, 3445.

106. Metzner, Ralph (1993). Letter from Ralph Metzner. Newsletter of the Multidisciplinary Association for Psychedelic Studies MAPS. 4 (1). Retrieved 8 January 2015.
107. Miller, W.R., and Rollnick, S. (1991). Motivational Interviewing:
Preparing People To Change Addictive Behavior. New York: Guilford Press.
108. Miller WR, Wilbourne PL. (2002) Mesa Grande: a methodological analysis of clinical trials of treatments for alcohol use disorders. Addiction. 2002; 97:265-277 109. Mitchell, J.M., Bogenschutz, M., Lilienstein, A. et al. MDMA-assisted therapy for severe PTSD: a randomized, double-blind, placebo-controlled phase 3 study. Nat Med 27, 1025-1033 (2021). https://doi.org/10.1038/s41591-021-01336-3.
110. Mithoefer MC, Wagner MT, Mithoefer AT, Jerome L, Martin SF, Yazar-Klosinski B, Michel Y, Brewerton TD, Doblin R. (2013) Durability of improvement in post-traumatic stress disorder symptoms and absence of harmful effects or drug dependence after 3,4-methylenedioxymethamphetamine-assisted psychotherapy: a prospective long-term follow-up study. Journal of Psychopharmacology. 27(1):28-39.
111. Mithoefer MC, Wagner TM, Mithoefer AT, Jerome L, Doblin R (2010) The safety and efficacy of 3,4-methylenedioxymethamphetamine-assisted psychotherapy in subjects with chronic, treatment-resistant posttraumatic stress disorder: the first randomized controlled pilot study. Journal of Psychopharmacology.25(4): 439-452.
112. Mithoefer MC (2017). A Manual for MDMA-Assisted Psychotherapy in the Treatment of Posttraumatic Stress Disorder. Ver. 8.1 https://maps.org/research-archive/mdma/MDMA-Assisted-Psychotherapy-Treatment-Manual-Version7-19Aug15-FINAL.pdf.
113. Mithoefer, M.C., Feduccia, A.A., Jerome, L. etal. MDMA-assisted psychotherapy for treatment of PTSD: study design and rationale for phase 3 trials based on pooled analysis of six phase 2 randomized controlled trials. Psychophannacology 236, 2735-2745 (2019).
https ://doi . org/10.1007/s00213 -019-05249-5.
114. Moos, R. 1-1., & Moos, B. S. (2006). Rates and predictors of relapse after natural and treated remission from alcohol use disorders. Addiction (Abingdon, England), /0/(2), 212-222.
doi:10.1110.1360-0443.2006.01310.x).
115. Morisky DE, Green LW, Levine DM. Concurrent and predictive validity of a self-reported measure of medication adherence. Med Care. 1986 Jan;24(1):67-74. doi:
10.1097/00005650-198601000-00007. P1V11D: 3945130.
116. Mundt, JC et al. (2013). "Prediction of suicidal behavior in clinical research by lifetime suicidal ideation and behavior ascertained by the electronic Columbia-Suicide Severity Rating Scale.".The Journal of clinical psychiatry 74 (9): 887-93.
117. Multidisciplinary Association for Psychedelic Studies (MAPS) (2016) Investigators Brochure for MDMA-Assisted Psychotherapy. Available for download at:
https://maps.org/research/mdma/mdma-research-timeline/104-other-mdma-resources/5400-mdma-investigator-s-brochure-and-fda-annual-report&Itemid=485 118. Nair JB, Hakes L, Yazar-Klosinski B, Paisner K, Fully Validated, Multi-Kilogram cGMP
Synthesis of MDMA, ACS Omega (IF3.512), 2021-12-20 10.1021/acsomega.1c05520.

119. Nash JF, Roth BL, Brodkin JD, et al. (1994) Effect of the R(-) and S(+) isomers of MDA
and MDMA on phosphatidyl inositol turnover in cultured cells expressing 5-HT2A
or 5-HT2C receptors. Neurosci Lett 1994;177:111-5.
120. National Clinical Guideline Centre (2010). 2 Acute Alcohol Withdrawal.
Alcohol Use Disorders: Diagnosis and Clinical Management of Alcohol-Related Physical Complications (No. 100 ed.). London: Royal College of Physicians (UK). Archived from the original on 31 January 2014.
121. National Collaborating Centre for Mental Health (UK). Alcohol-Use Disorders: Diagnosis, Assessment and Management of Harmful Drinking and Alcohol Dependence.
Leicester (UK): British Psychological Society; 2011. (NICE Clinical Guidelines, No.
115.) 2, ALCOHOL DEPENDENCE AND HARMFUL ALCOHOL USE. Available from:
https://www.ncbi.nlm.nih.gov/books/NBK65500/
122. National Institute for Health and Care Excellence, NICE (June 2013) Alcohol-use disorder diagnosis and clinical management of alcohol-related physical complications 123. Neff, KD. (2003). Development and validation of a scale to measure self-compassion. Self and Identity, 2, 223-250.
124. NICE (2011) Alcohol-use Disorders: Diagnosis, Assessment and Management of Harmful Drinking and Alcohol Dependence. NICE clinical guideline 115. Available at www.nice.org.uk/CG115 ["NICE Guidelines-]
125. Nichols, D.E. (1986). "Differences Between the Mechanism of Action of MDMA, MBDB, and the Classic Hallucinogens. Identification of a New Therapeutic Class:
Entactogens". J.
Psychoactive Drugs 18(4): 305-13.
126. Oehen, P., Traber, R., Widmer, V., & Schnyder, U. (2013). A randomized, controlled pilot study of MDMA ( 3,4-Methylenedioxymethamphetamine)-assisted psychotherapy for treatment of resistant, chronic Post-Traumatic Stress Disorder (PTSD). Journal of psychopharmacology (Oxford, England), 27(1), 40-52.
https://doi.org/10.1177/0269881112464827.
127. Oehen P. Traber, R., Widmer, V., Schnyder, U. (2013) Pilot study of MDMA-assisted psychotherapy for treatment-resistant PTSD. J Psychopharmacol; 27(1): 40-52.
128. Oeri H. E. (2021). Beyond ecstasy: Alternative entactogens to MDMA with potential applications in psychotherapy. J psychopharmacol., 35(5), 512-536.
129. Office for National Statistics (ONS). (7 December 2021). Alcohol-specific deaths in the UK: registered in 2020. Available at (last accessed 6 January 2022):
https://www.ons.gov.uk/peoplepopulationandcommunity/healthandsocialcare/causeso fdeath /bulletins/al coholrelateddeathsintheunitedkingdom/regi steredin 2020 130. Osaji, J., Ojimba, C., & Ahmed, S. (2020). The Use of Acceptance and Commitment Therapy in Substance Use Disorders: A Review of Literature. Journal of clinical medicine research, 12(10), 629-633. https://doi.org/10.14740/jocmr4311.
131. Paille, F. and Martini, H. (2014). "Nalmefene: a new approach to the treatment of alcohol dependence". Substance Abuse and Rehabilitation 5 (5): 87-94.
132. Parrott and Hindmarch, (1980). The Leeds Sleep Evaluation Questionnaire in Psychopharmacological Investigations -- a Review. Psychopharmacology 71, 173-179.

133. Passie (2012) Healing with Entactogens: Therapist and Patient Perspectives on MDMA-Assisted Group Psychotherapy (Ben Lomond, CA: Multidisciplinary Association for Psychedelic Studies).
134. Paterson et al. (2015) The Imperial College Cambridge Manchester (ICCAM) platform study: an experimental medicine platform for evaluating new drugs for relapse prevention in addiction. Part A: study description. J Psychopharmacol, September 2015, vol. 29, no. 9, 135. Piano M. R. (2017). Alcohol's Effects on the Cardiovascular System.
Alcohol research current reviews, 38(2), 219-241.
136. Pitts, E. G., Curry, D. W., Hampshire, K. N., Young, M. B., & Howell, L.
L. (2018). ( )-MDMA and its enantiomers: potential therapeutic advantages of R(-)-MDMA.
Psychopharmacology, 235(2), 377-392. https://doi.org/10.1007/s00213-017-4812-5.
137. Project MATCH Research Group (1998a). Matching alcoholism treatments to client heterogeneity: Project MATCH three year drinking outcomes. Alcoholism:
Clinical and Experimental Research, 22,1300-1311.
138. Rehm J. The risks associated with alcohol use and alcoholism. Alcohol Res Health.
2011;34(2): 135-43 139. Rezvani, A.H., Garges, P.L., Miller, D.B. and Gordon C.J. (1992) Attenuation of Alcohol Consumption by MDMA (Ecstasy) in Two Strains of Alcohol-preferring Rats.
Pharmacology, Biochemistry and Behaviour, Vol 43, pp. 103-110.
140. Rogers G, Elston J, Garside R, et al. (2009) The harmful health effects of recreational ecstasy: a systematic review of observational evidence. Health Technol Assess 2009; 13(6):
iii-iv, ix-xii, 1-315.
141. Wisner S, Hackl-Herrwerth A, Leucht S, Lehert P. Vecchi S. Soyka M
(2010). Wisner, Susanne, ed. "Acamprosate for alcohol dependence".Cochrane Database of Systematic Reviews (9): CD004332.doi:10.1002/14651858.CD004332.pub2.
142. Rydzynski, Z., Cwynar, S., Grzelak, L., 1968. Preliminary report on the experience with psychotomimetic drugs in the treatment of alcoholism. Act. Nerv. Super.
(Praha) 10 (3), 273.
143. Saitz M, Mayo-Smith ME, Redmond HA, Bernard DR, Calkins DR. (1994) Individualized treatment for alcohol withdrawal. A randomized double-blind controlled trial.
JAMA
1994;272:519-23.
144. Saleemi, S., Pennybaker, S. J., Wooldridge, M., & Johnson, M. W. (2017).
Who is 'Molly'?
MDMA adulterants by product name and the impact of harm-reduction services at raves.
Journal of psychopharmacology (Oxford, England), 31(8), 1056-1060.
145. Sayette, M. A., Shiffman, S., Tiffany, S. T., Niaura, R. S., Martin, C.
S., & Shade!, W. G.
(2000). The measurement of drug craving. Addiction (Abingdon, England), 95 Suppl 2(Suppl 2), S189-S210. hftps://doi.org/10.1080/09652140050111762 146. Schmid, Gasser, Oehen, & Liechti (2020). Acute subjective effects in LSD-and MDMA-assisted psychotherapy. Journal of psychopharmacology, 0269881120959604.

147. Selvaraj S, Hoshi R, Bhagwagar Z, et al. (2009) Brain serotonin transporter binding in former users of MDMA ('ecstasy'). Br J Psychiatry; 194(4): 355-9.
148. Sessa, B. (2012) Could MDMA be useful in the treatment of PTSD? Progress in Neurology and Psychiatry. Volume 15, Issue 6, pages 4-7 149. Sessa & Fischer (2015), Underground MDMA-, LSD-and 2-CB-assisted individual and group psychotherapy in Zurich: Outcomes, implications and commentary. Drug Science, Policy and Law, 2,2050324515578080.
150. Sessa B. (2017) Why MDMA therapy for alcohol use disorder? And why now?
Neuropharmacol. 142:83-88. doi: 10.1016/j.neuropharm.2017.11.004. PM1D:
29126911.
151. Sessa, B., Higbed, L. and Nutt, DJ (2019) A Review of 3,4-methylenedioxymethamphetamine (MDMA)-Assisted Psychotherapy. Frontiers Psychiatry 10:138. doi: 10.3389/fpsyt.2019.00138 152. Sessa, B., Sakal, CM., O'Brien, S. & Nutt, D.J. (2019) First study of safety and tolerability of MDMA-assisted psychotherapy in patients with alcohol use disorder:
preliminary data on the first four participants. MDMA-AUD ¨ BMJ Case Reports paper ¨ July 2019(1) BMJ
Case Reports. 2019;12:e230109 153. Sessa, B. et al. (2020). How well are Patients doing Post-Alcohol Detox in Bristol? Results from the Outcomes Study. J. Alcohol Drug Depend. Subst. Abuse, 6:021 154. Sessa, B., Higbed, L., O'Brien, S., Durant, C., Sakai, C., Titheradge, D., Williams, T. M., Rose-Morris, A., Brew-Girard, E., Burrows, S., Wiseman, C., Wilson, S., Rickard, J., &
Nutt, D. J. (2021). First study of safety and tolerability of 3,4-methylenedioxymethamphetamine-assisted psychotherapy in patients with alcohol use disorder. Journal of psychopharmacology (Oxford, England), 35(4), 375-383.
https://doi.org/10.1177/0269881121991792 155. Shapiro SL, et al. Mechanisms of mindfulness. Journal of Clinical Psychology.
2006;62(3): 373-386.
156. Sheehan DV, Lecrubier Y, Sheehan KH, Amorim P, Janays J, Weiller E, Hergueta T, Baker R, Dunbar GC. (1998) The Mini-International Neuropsychiatric Interview (M.I.N.I.): the development and validation of a structured diagnostic psychiatric interview for DSM-IV
and ICD-10. J Clin Psychiatry.;59 Suppl 20:22-33;quiz 34-57.
157. Sheth et al. (1980). Compressed tablets, in Pharm. dosage forms, Vol. 1, Lieberman &
Lachtman, eds., Dekker, NY.
158. Shulgin, A. T. (1986). The Background and Chemistry of MDMA. Journal of Psychoactive Drugs, 18(4), 291-304. doi:10.1080/02791072.1986.1047.
159. Singh et al. Encyclopedia of Pharmaceutical Technology, 2nd Ed., pp. 751-753 (2002).
160. Sloshower, J., Guss, J., Krause, R., Wallace, R.M., Williams, M.T., Reed, S., Skinta., M. D.
(2020). Psilocybin-assisted therapy of major depressive disorder using Acceptance and Commitment Therapy as a therapeutic frame. Journal of Contextual Behavioral Science, 15;
12-19.

161. Sobel!, L.C., Agrawal, S., Annis, H.M., etal. (2001). Cross-cultural evaluation of two drinking related assessment instruments: Alcohol Timeline Follow Back and Inventory of Drinking Situations. Substance Use & Misuse, 36, 313-331.
162. Sobel! L C, Sobel! MB, 1996, Timeline FollowBack user's guide: A calendar method for assessing alcohol and drug use. Addiction Research Foundation, Toronto, Ontario, Canada;
J. Anal. Toxicolo., 2002, 26: 393-400.
163. Sobel! LC, Sobel! MB, Maisto SA. Follow-up attrition in alcohol treatment studies: is 'no news' bad news, good news or no news? Drug Alcohol Depend. 1984 Jan;13(1):1-7.
164. Soyka M, Horak M. Outpatient alcohol detoxification: implementation efficacy and outcome effectiveness of a model project. Eur Addict Res. 2004;10(4):180-7.
165. Soyka M, Wisner S. (2008). "Opioid antagonists for pharmacological treatment of alcohol dependence - a critical review".Curr Drug Abuse Rev 1 (3): 280-91.
166. Spitzer R.L., Kroenke K., Williams J.B., Lowe B. (2006) A brief measure for assessing generalized anxiety disorder: the GAD-7. Arch intern med. 22;166(10):1092-7.
167. Stanford Sparqtools: Health-Related Quality of Life Scale. Retrieved January 6, 2022 from sparqtools.org/mobility-measure/health-related-quality-of-life-scale/#all-survey-questions 168. Stockwell T, Murphy D, Hodgson R. (1983) The severity of alcohol dependence questionnaire: its use, reliability and validity. Bri. Journal of Addictions;78:145-155.
169. Stolaroff M. (2004) The Secret Chief Revealed: Conversations with a pioneer of the underground therapy movement. Chapter 2, pages 45-49. Published by:
Multidisciplinary Association for Psychedelic Studies, Sarasota, USA.
170. Sullivan JT, Sykora K, Schneiderman J, Naranjo CA, Sellers EM. Assessment of alcohol withdrawal: the revised clinical institute withdrawal assessment for alcohol scale (CIWA-Ar). Br J Addict. 1989 Nov;84(11):1353-7.
171. Sumnall, H. R, Cole, J. and Jerome, L. (2006) The varieties of ecstatic experience: an exploration of the subjective experiences of ecstasy. Journal of Psychopharmacology. 20(5) (2006) 670-682.
172. Teasdale, J. D., Segal, Z. V., Williams, J. M., Ridgeway, V. A., Soulsby, J. M., & Lau, M.
A. (2000). Prevention of relapse/recurrence in major depression by mindfulness-based cognitive therapy. Journal of consulting and clinical psychology, 68(4), 615-623.
https://doi.org/10.1037//0022-006x.68.4.615.
173. Thompson MR, Callaghan PD, Hunt GE, Cornish JL, McGregor IS. (2007) A
role for oxytocin and 5-HT(1A) receptors in the prosocial effects of 3,4 methylenedioxymethamphetamine ("ecstasy"). Neuroscience; 146(2): 509-14.
174. Ugochukwu, C, Bagot, KS; Delaloye, S, Pi, S, Vien, L; Garvey, T, Bolotaulo, Nestor I, Kumar, N, IsHak, WW. (2013). The Importance of Quality of Life in Patients with Alcohol Abuse and Dependence, Harvard Review of Psychiatry: 21(1):1-17.
175. UKATT Research Team (2005). Effectiveness of treatment for alcohol problems: Findings of the randomised UK Alcohol Treatment Trial (UKATT). British Med. J., 311, 541-544.

176. U.S. Department of Health and Human Services. (2021). What is a standard drink?
National Institute on Alcohol Abuse and Alcoholism. Retrieved Dec. 30, 2021, from niaaa.nih.gov/alcohols-effects-health/overview-alcohol-consumption/what-standard-drink 177. Villalobos-Gallegos, L., Perez-Lopez, A., Mendoza-Hassey, R., Graue-Moreno, J., &
Marin-Navarrete, R. (2015). Psychometric and diagnostic properties of the Drug Abuse Screening Test (DAST): Comparing the DAST-20 vs. the DAST-10. Salud Mental, 38(2), 89-94. https://doi.org/10.17711/SM.0185-3325.2015.012 178 Ware, JF, Jr, Sherhourne CD (1992) The MOS 36-item short-form health survey (SF-36) T
Conceptual framework and item selection. Med Care. 30(6):473-83. PMID:
1593914.
179. Ware, JE. (1993) Health survey manual and interpretation guide. Section 6: Scoring the SF-36. The Health Institute, New England Medical Center. Nimrod Press, Boston, Massachusetts.
180. Watson, L. and Beck, J. (1991) New Age Seekers: MDMA as an Adjunct to Spiritual Pursuit. Journal of Psychoactive Drugs. Vol 23(3), Jul-Sep 1991.
181. Watts, R., & Luoma, J. (2020). The use of the psychological flexibility model to support psychedelic assisted therapy. Journal of contextual behavioral science, 15, 92-102.
182. The WHOQoL GROUP (1995) The World Health Organization Quality Of Life Assessment (WHOQoL): Position Paper From the World Health Organization. Soc Sci Med1014031409 183. WHO. (1996). WHOQOL-BREF: Introduction, administration, scoring and generic version of the assessment, Programme on Mental Health, World Health Organization, Geneva, 184. Witkiewitz K, et al., Recommendations for the Design and Analysis of Treatment Trials for Alcohol Use Disorders. Alcohol Clin Exp Res. 2015 Sep;39(9):1557-70.
185. Witkiewitz, K., Litten, R. Z., & Leggio, L. (2019). Advances in the science and treatment of alcohol use disorder. Science Advances, 5(9).
https://doi.org/10.1126/sciadv.aax4043 186. Yehuda, R., et al. (2016). Holocaust Exposure Induced Intergenerational Effects on FKBP5 Methylation. Biological psychiatry, 80(5), 372-380.
187. Yi, H., Shin, K. and Shin, C. (2006), Development of the Sleep Quality Scale. Journal of Sleep Research, 15: 309-316.
188. Yin, S., Njai, R., Barker, L., Siegel, P. Z., & Liao, Y. (2016).
Summarizing health-related quality of life (HRQOL): development and testing of a one-factor model.
Population health metrics, 14, 22. https://doi.org/10.1186/s12963-016-0091-3 189. Yubero-Lahoz, S., et al. Sex Differences in 3,4 methylenedioxymethamphetamine (MDMA;
Ecstasy)-Induced Cytochrome P450 2D6 Inhibition in Humans. Clin Pharmacokinet, 2011.
50(5): p. 319-29.
190. Zanger, U. M., & Schwab, M. (2013). Cytochrome P450 enzymes in drug metabolism:
regulation of gene expression, enzyme activities, and impact of genetic variation.
Pharmacology & therapeutics, 138(1), 103-141.
https://doi.org/10.1016/j.pharmthera.2012.12.007.

Claims (205)

1. A method of treating alcohol use disorder (AUD) in a subject diagnosed therewith, comprising facilitating an MDMA-assisted psychotherapy regimen, wherein the regimen comprises one or more MDMA-assisted psychotherapy sessions and one or more non-drug psychotherapy sessions.
2. The method of claim 1, further comprising an alcohol detoxification course prior to the MDMA-assisted psychotherapy regimen
3. The method of claim 2, further comprising a pre-detoxification motivational group therapy course prior to the alcohol detoxification course.
4. The method of claim 1, further comprising a group therapy preparation course prior to the MDMA-assisted psychotherapy regimen.
5. A method of maintaining reduced alcohol intake in a subject diagnosed with AUD, comprising facilitating an MDMA-assisted psychotherapy regimen, wherein the regimen comprises one or more MDMA-assisted psychotherapy sessions and one or more non-drug psychotherapy sessions.
6. The method of claim 5, further comprising an alcohol detoxification course prior to the MDMA-assisted psychotherapy regimen.
7. The method of claim 6, further comprising a pre-detoxification motivational group therapy course prior to the alcohol detoxification course.
8. The method of claim 5, further comprising a group therapy preparation course prior to the MDMA-assisted psychotherapy regimen.
9. The method of any of claims 5-8, wherein the reduced alcohol intake is maintained for at least 1 month after the subject has completed the MDMA-assisted psychotherapy regimen.
1 0 The method of any of claims 5-8, wherein the reduced alcohol intake is maintained for at least 3 months after the subject has completed the MDMA-assisted psychotherapy regimen.
11. The method of any of claims 5-8, wherein the reduced alcohol intake is maintained for at least 6 months after the subject has completed the MDMA-assisted psychotherapy regimen.
12. The method of any of claims 5-8, wherein the reduced alcohol intake is maintained for at least 9 months after the subject has completed the MDMA-assisted psychotherapy regimen.
13. The method of any of claims 5-8, wherein the reduced alcohol intake is maintained for at least 12 months after the subject has completed the MDMA-assisted psychotherapy regimen.
14. The method of any of claims 5-13, wherein alcohol intake is reduced by at least 50%.
15. The method of claim 14, wherein alcohol intake is reduced by at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 100%.
16. The method of claim 15, wherein the subject remains completely abstinent from alcohol.
17. A method of preventing relapse into alcohol use in a subject diagnosed with AUD, comprising facilitating an MDMA-assisted psychotherapy regimen, wherein the regimen comprises one or more MDMA-assisted psychotherapy sessions and one or more non-drug psychotherapy sessions.
18. The method of claim 17, further comprising an alcohol detoxification course prior to the MDMA-assisted psychotherapy regimen.
19. The method of claim 18, further comprising a pre-detoxification motivational group therapy course prior to the alcohol detoxification course.
20. The method of claim 17, further comprising a group therapy preparation course prior to the MDMA-assisted psychotherapy regimen.
21. The method of any of claims 17-20, wherein the relapse into alcohol use is prevented for at least 1 month after the subject has completed the MDMA-assisted psychotherapy regimen.
22. The method of any of claims 17-20, wherein the relapse into alcohol use is prevented for at least 3 months after the subject has completed the MDMA-assisted psychotherapy regimen.
23. The method of any of claims 17-20, wherein the relapse into alcohol use is prevented for at least 6 months after the subject has completed the MDMA-assisted psychotherapy regimen.
24. The method of any of claims 17-20, wherein the relapse into alcohol use is prevented for at least 9 months after the subject has completed the MDMA-assisted psychotherapy regimen.
25. The method of any of claims 17-20, wherein the relapse into alcohol use is prevented for at least 12 months after the subject has completed the MDMA-assisted psychotherapy regimen.
26. A method of treating the symptoms of a comorbid psychiatric disorder in a subject diagnosed with AUD, comprising facilitating an MDMA-assisted psychotherapy regimen, wherein the regimen comprises one or more MDMA-assisted psychotherapy sessions and one or more non-drug psychotherapy sessions.
27. The method of claim 26, further comprising an alcohol detoxification course prior to the MDMA-assisted psychotherapy regimen
28. The method of claim 27, further comprising a pre-detoxification motivational group therapy course prior to the alcohol detoxification course.
29. The method of claim 26, further comprising a group therapy preparation course prior to the MDMA-assisted psychotherapy regimen.
30. The method of any of claims 26-29, wherein the comorbid psychiatric disorder is any of post-traumatic stress disorder (PTSD), adjustment disorder, affective disorder, depression, atypical depression, postpartum depression, catatonic depression, a depressive disorder due to a medical condition, premenstrual dysphoric disorder, seasonal affective disorder, dysthymia, anxiety, phobia disorders, binge disorders, body dysmorphic disorder, a mood disorder related to another health condition, disruptive behavior disorders, eating disorders, impulse control disorders, obsessive compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), personality disorders, attachment disorders, dissociative disorders, and sub stance-related disorders including substance use disorder (SUD).
31. The method of claim 30, wherein the comorbid psychiatric disorder is depression or anxiety.
32 The method of claim 3 1 , wherein the comorbid psychiatric di sorder is depression, and the symptoms are treated as demonstrated by a reduction in a PHQ-9 depression score.
33. The method of claim 31, wherein the comorbid psychiatric disorder is anxiety, and the symptoms are treated as demonstrated by a reduction in a GAD-7 anxiety score.
34. The method of any of claims 26-29, wherein the comorbid psychiatric disorder is a behavioral addiction, including any of pathological gambling, kleptomania, pyromania, compulsive buying, compulsive sexual behaviour, Internet addiction, pornography, and binge eating disorder.
35. The method of any of claims 1-4, wherein the subject has dependent use AUD.
36. The method of any of claims 1-4, wherein the subject has harmful use AUD.
37. The method of any of claims 1-4, wherein the subject has alcohol abuse, alcohol dependence, alcohol addiction, or alcoholism.
38. The method of any of claims 1-4, wherein the subject has demonstrated motivation to reduce their alcohol intake.
39. The method of either of claims 2 or 3, wherein the subject has successfully completed the alcohol detoxification course prior to the MDMA-assisted psychotherapy regimen.
40. The method of either of claims 3 or 39, wherein the subject has successfully completed the pre-detoxification motivational group therapy course prior to the alcohol detoxification course.
41. The method of claim 4, wherein the subject has successfully completed the group therapy preparation course prior to the MDMA-assisted psychotherapy regimen.
42. The method of any of claims 1-4, wherein the subject is diagnosed with AUD as defined by a version of the Diagnostic and Statistical Manual of Mental Disorders (DSM), comprising scoring above the diagnostic threshold for AUD.
43. The method of claim 42, wherein severity of AUD is determined using one or more questionnaires.
44. The method of claim 43, wherein the one or more questionnaire(s) comprise the Severity of Alcohol Questionnaire (SADQ) and/or the Short Inventory of Problems for Alcohol (SIP) questionnaire.
45. The method of claim 44, wherein the one or more questionnaire(s) comprise the Severity of Alcohol Questionnaire (SADQ).
46. The method of claim 45, wherein the severity of AUD is scored on the SADQ as mild (score of less than 16), moderate (score of 16-30), or severe (score of greater than or equal to 31).
47. The method of claim 46, wherein the severity of AUD is mild (score of less than 16).
48. The method of claim 46, wherein the severity of AUD is moderate (score of 16-30).
49. The method of claim 46, wherein the severity of AUD is severe (score of greater than or equal to 31).
50. The method of any of claims 1-4, wherein said method results in one or more of: (a) a reduction of alcohol use, (b) a reduction of alcohol cravings, (c) a promotion of alcohol abstinence, (d) a prevention of relapse into alcohol use, or (e) an improvement of at least one symptom of alcohol use disorder.
51 The method of any of claims 1-4, wherein said method results in one or more of:
(a) an increase in quality of life, (b) an increase in psychosocial functioning, (c) a decrease in use or frequency of a prescription medication, (d) a decrease in use or frequency of a recreational drug, (e) a decrease in obsessive compulsive thoughts, (f) a decrease in suicidality, (g) an increase in feelings of empathy, or (h) an increase in self-compassion.
52. The method of any of claims 1-4, wherein said method results in one or more of:
(a) an increase in physical functioning; (b) a decrease in role limitations due to physical health; (c) an increase in emotional well-being; (d) a decrease in role limitations due to emotional problems; (e) an increase in energy; (f) a decrease in fatigue; (g) an increase in social functioning; (h) a decrease in pain; or (i) an increase in general health.
53. The method of claim 52 wherein the increase and/or the decrease is demonstrated by a patient-reported measure on a Short Form (36) Health Survey.
54. The method of any of claims 50-53, wherein the results are measured at least 1 month, at least 3 months, at least 6 months, at least 9 months, or at least 1 year from the end of detoxification, if detoxification is completed, from baseline, from the first psychotherapy session, or from the completion of the MDMA-assisted psychotherapy regimen.
55. The method of any of claims 1-54, wherein the MDMA-assisted psychotherapy regimen comprises one or more non-drug psychotherapy sessions prior to and after the one or more MDMA-assisted psychotherapy sessions.
56. The method of claim 55, wherein the MDMA-assisted psychotherapy regimen comprises more than one non-drug psychotherapy session prior to and after the one or morc MDMA-assisted psychotherapy sessions.
57. The method of any of claims 1-55, wherein the MDMA-assisted psychotherapy regimen compri ses administering one or more doses of MDMA during the one or more MDMA-assisted psychotherapy sessions.
58. The method of any of claims 1-57, wherein the MDMA-assisted psychotherapy regimen comprises repeated MDMA-assisted psychotherapy sessions.
59. The method of any of claims 1-58, wherein the MDMA-assisted psychotherapy regimen comprises a first MDMA-assisted psychotherapy session and a subsequent MDMA-assisted psychotherapy session.
60. The method of claim 59, wherein the MDMA-assisted psychotherapy regimen comprises one, two, three, or four non-drug psychothetapy sessions prior to the first MDMA-assisted psychotherapy session.
61. The method of either claim 59 or 60, wherein the MDMA-assisted psychotherapy regimen comprises two non-drug psychotherapy sessions prior to the first MDMA-assisted psychotherapy session.
62. The method of any of claims 59-61, wherein the MDMA-assisted psychotherapy regimen comprises one or more non-dnig psychotherapy session(s) after the first MDMA-assisted psychotherapy session.
63. The method of any of claims 59-62, wherein the MDMA-assisted psychotherapy regimen comprises one, two, three, four, five, or six non-drug psychotherapy sessions after the first MDMA-assisted psychotherapy session and prior to a subsequent MDMA-assisted psychotherapy session.
64. The method of claim 63, wherein the MDMA-assisted psychotherapy regimen comprises three non-drug psychotherapy sessions after the first MDMA-assisted psychotherapy session and prior to the subsequent MDMA-assisted psychotherapy session.
65. The method of claim 63, wherein the subsequent MDMA-assisted psychotherapy session is a second, a third, a fourth, or a fifth MDMA-assisted psychotherapy session.
66. The method of 65, wherein the subsequent MDMA-assisted psychotherapy session is the second MDMA-assisted psychotherapy session.
67. The method of any of claims 1-66, wherein the MDMA-assisted psychotherapy regimen comprises two MDMA-assisted psychotherapy sessions and eight non-drug psychotherapy sessions.
68. The method of any of claims 1-66, wherein the MDMA-assisted psychotherapy regimens comprises three MDMA-assisted psychotherapy sessions and 11 psychotherapy non-drug sessions.
69. The method of any of claims 1-68, wherein the one or more MDMA-assisted psychotherapy sessions comprise administering one single dose of MDMA.
70. The method of claim 59, wherein the first and the subsequent MDMA-assisted psychotherapy sessions each comprise administering one single dose of MDMA.
71. The method of claim 59-68, wherein the one or more MDMA-assisted psychotherapy sessions comprise administering two doses of MDMA, an initial dose and a booster dose.
72. The method of claims 59-68 and 71, wherein the first and the subsequent MDMA-assisted psychotherapy sessions each comprise two doses of MDMA, an initial dose and a booster dose.
73. The method of either of claims 71 or 72, wherein the booster dose is less than the initial dose.
74 The method of claim 73, wherein the booster dose is about 10% to about 90% of the amount of the initial dose.
75. The method of either of claims 73 or 74, wherein the booster dose is about 25%, about 50%, or about 75% of the amount of the initial dose.
76. The method of either of claims 73 or 74, wherein the booster dose is about 10%, 15%, 20%, 25%, 30%, 35%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, or 90% of the amount of the initial dose.
77. The method of any of claims 71-76, comprising administering the booster dose about 30 minutes, one hour, two hours, three hours, or four hours after the initial dose.
78. The method of any of claims 71-76, comprising administering the booster dose two hours following the initial dose, or at such time as to extend peak subjective effects or to optimize the plasma concentration-time profile.
79. The method of any of claims 1-66, wherein the MDMA-assisted psychotherapy regimen comprises between 5 and 20 psychotherapy sessions, inclusive, during which at least two of said psychotherapy sessions the subject is administered MDMA.
80. The method of any of claims 1-79, comprising administering a total dose of about 40 mg to 200 mg of MDMA, wherein the range is inclusive.
81. The method of any of claims 1-79, comprising administering a total dose of about 40 mg to 100 mg, about 100 mg to 150 mg, about 150 to 200 mg, about 175 mg to 200 mg, about 180 mg to 190 mg, about 180 to 200 mg, or about 185 to 200 mg of MDMA, wherein each range is inclusive.
82. The method of any of claims 1-79, comprising administering a total dose of about 180 mg, 181 mg, 182 mg, 183 mg, 184 mg, 185 mg, 186 mg, 187 mg, 188 mg, 189 mg, or 190 mg of MDMA.
83. The method of any of claims 1-79, comprising administering a total dose of about 187.5 mg of MDMA.
84. The method of any of claims 1-83, comprising administering MDMA as a pharmaceutically acceptable salt, as a free base, or as a polymorph of MDMA.
85. The method of any of claims 1-84, comprising administering MDMA as a racemate.
86. The method of any of claims 1-84, comprising administering MDMA as an enantiomerically enriched mixture of R-MDMA and S-MDMA having an enantiomeric excess in any amount greater than 0%.
87. The method of any of claims 1-84, comprising administering MDMA as a pure or substantially pure individual enantiomer of R-MDMA or S-MDMA having an enantiomeric excess greater than 90%.
88. The method of any of claims 1-87, wherein the psychotherapy in MDMA-assisted and non-drug sessions comprises motivational interviewing.
89. The method of any of claims 1-87, wherein the psychotherapy in MDMA-assisted and non-drug sessions comprises cognitive behavioral therapy.
90. The method of any of claims 1-87, wherein the psychotherapy in MDMA-assisted and non-drug sessions comprises motivational interviewing and cognitive behavioral therapy.
91. The method of either of claims 89 or 90, wherein the cognitive behavioral therapy is a modified form of cognitive behavioral therapy comprising elements of Acceptance and Commitment Therapy (ACT).
92. The method of any of claims 1-91, wherein the MDMA-assisted and drug-free psychotherapy sessions comprise one or more of:
(a) supporting subjects to maintain and/or improve a reduction in alcohol consumption;
(b) preventing relapse into heavy drinking; and (c) developing skills to lead a fulfilling life without problematic drinking.
93. A method of selecting a subject for MDMA-assisted psychotherapy, comprising identifying a subject who meets one or more of the following criteria:
(a) has an AUD diagnosis, wherein the severity of AUD is mild, moderate, or severe;
(b) has completed detoxification of alcohol prior to administration of MDMA-assisted psychotherapy; and (c) has tapered off of medications known to interact with or attenuate the effects of MDMA prior to administration of MDMA-assisted psychotherapy;
wherein the subject does not experience adverse events associated with administration of MDMA.
94. A method of minimizing or eliminating the risk of adverse events related to MDMA-assisted psychotherapy in a subject having AUD comprising:
(a) selecting a subject who has been diagnosed with AUD, wherein the severity of AUD is mild, moderate, or severe;
(b) facilitating detoxification of alcohol prior to administration of MDMA-assisted psychotherapy; and (c) tapering off medications known to interact with or attenuate the effects of MDMA prior to administration of MDMA-assisted psychotherapy;
wherein the subject does not experience adverse events associated with administration of MDMA.
95. The method of either of claims 93 or 94, wherein detoxification comprises gradually cutting down alcohol consumption over many weeks and/or is a medically assisted detoxification regime.
96. The method of any of claims 93-95, further comprising facilitating a pre-detoxification motivational group therapy course prior to facilitating the detoxification of alcohol.
97. A method of selecting a subject for MDMA-assisted psychotherapy, comprising identifying a subject who meets one or more of the following criteria:
(a) has an AUD diagnosis, wherein the severity of AUD is mild, moderate, or severe;
(b) has completed a group therapy preparation course prior to administration of 1VIDMA-assisted psychotherapy; and (c) has tapered off of medications known to interact with or attenuate the effects of MDMA prior to administration of MDMA-assisted psychotherapy;
wherein the subject does not experience adverse events associated with administration of MDMA.
98. The method of any of claims 1-97, comprising monitoring the subject having AUD
for adverse events during administration of MDMA.
99. The method of claim 98, wherein the adverse events during administration of MDMA
comprise one or more of hyperthermia, hypertension, and elevated heart rate.
100. The method of either of claims 98 or 99, wherein the subject diagnosed with AUD
does not display adverse events during administration of MDMA.
101. The method of any of claims 1-100, comprising monitoring the subject having AUD
for adverse events after administration of MDMA.
102. The method of claim 101, wherein the adverse events after administration of MDMA
comprise mood disturbances and/or depressed affect.
103. The method of claim 102, wherein the subject does not report a disturbance in mood or a depressed affect after administration of MDMA-assisted psychotherapy.
104. The method of claim 103, wherein the subject does not report a disturbance in mood or a depressed affect within about one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, or ten days after administration.
105. The method of any of claims 101-104, wherein the subject diagnosed with AUD does not display adverse events after administration of MDMA-assisted psychotherapy.
106. The method of any of claims 1-100, comprising monitoring the subject having AUD
for adverse events both during and after administration of MDMA.
107. The method of claim 106, wherein the adverse events during administration of MDMA comprise one or more of hyperthermia, hypertension, and elevated heart rate, and the adverse events after administration of MDMA comprise mood disturbances and/or depressed affect.
108. The method of either of claims 106 or 107, wherein the subject diagnosed with AUD
does not display or report adverse events during or after administration of MDMA-assisted psychotherapy.
109. The method of claim 108, wherein the subject does not display adverse events during administration of MDMA, or report adverse events after administration of MDMA, including within about one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, or ten days after administration.
110. The method of any of claims 1-109, comprising evaluating the severity of anxiety and/or depression in a subject diagnosed with AUD prior to MDMA-assisted psychotherapy.
111. The method of claim 110, wherein the severity of anxiety and/or depression are assessed using questionnaires.
112. The method of claim 111, wherein the severity of anxiety and/or depression are assessed using the Generalised Anxiety Disorder Assessment (GAD-7) and/or the Patient Health Questionnaire-9 (PHQ-9) questionnaire(s).
113. The method of claim 112, wherein the severity of anxiety and/or depression are also assessed after 1VIDMA-assisted psychotherapy, and are reduced after the 1VIDMA-assisted psychotherapy in comparison to levels preceding said MDMA-assisted psychotherapy.
114. The method of any of claims 1-113, wherein AUD is diagnosed as defined by a version of the Diagnostic and Statistical Manual of Mental Disorders (DSM), wherein diagnosis comprises scoring above the diagnostic threshold for AUD.
115. The method of any of claims 1-114, wherein the subject has dependent use AUD.
116. The method of any of claims 1-114, wherein the subject has harmful use AUD.
117. The method of any of claims 1-114, wherein the subject has alcohol abuse, alcohol dependence, alcohol addiction, or alcoholism.
118. The method of any of claims 1-117, wherein severity of AUD is determined using one or more questionnaires.
119. The method of claim 118, wherein the one or more questionnaire(s) comprise the Severity of Alcohol Questionnaire (SADQ) and/or the Short Inventory of Problems for Alcohol (SIP) questionnaire.
120. The method of claim 118 or claim 119, wherein the one or more questionnaire(s) comprise the Severity of Alcohol Questionnaire (SADQ).
121. The method of claim 120, wherein the severity of AUD is scored on the SADQ as mild (score of less than 16), moderate (score of 16-30), or severe (score of greater than or equal to 31).
122. The method of claim 121, wherein the severity of AUD is mild (score of less than 16).
123. The method of claim 121, wherein the severity of AUD is moderate (score of 16-30).
124. The method of claim 121, wherein the severity of AUD is severe (score of greater than or equal to 31).
125. The method of any of claims 1-124, which comprises an alcohol detoxification course prior to the MDMA-assisted psychotherapy regimen.
126. The method of any of claims 1-124, which comprises a pre-detoxification motivational group therapy course prior to the alcohol detoxification course.
127. The method of any of claims 1-124, which comprises a group therapy preparation course prior to the MDMA-assisted psychotherapy regimen.
128. The method of any of claims 1-127, wherein adverse events during and/or after administration of MDMA-assisted psychotherapy are not observed and/or reported.
129. A method of treating an alcohol use disorder (AUD) subject, the method comprising:
a. confirming the subject has alcohol use disorder, b. conducting a screening visit;
c. confirming the subject meets all inclusion criteria;
d. confirming the subject does not meet any exclusion criteria;
e. conducting a plurality of psychotherapy sessions as part of a treatment protocol, wherein at least three of the plurality of psychotherapy sessions include at least one dose of MDMA (MDMA-assisted psychotherapy sessions); and f. evaluating results at predetermined intervals after the subject completes the treatment protocol.
130. The method of claim 129, wherein the treatment protocol includes between 5 and 20 sessions or between 10 and 15 sessions.
131. The method of either of claims 129 or 130, wherein the treatment protocol includes 14 sessions.
132. The method of any of claims 129-131, wherein the treatment protocol includes 3 MDMA-assisted psychotherapy sessions, and 11 non-drug psychotherapy sessions.
133. The method of any of claims 129-132, wherein the MDMA-assisted psychotherapy sessions comprise at least one MDMA administration.
134. The method of claim 133, wherein the at least one MDMA administration comprises an initial dose and optionally a booster dose.
135. The method of claim 134, wherein the initial dose is between about 100 mg to about 150 mg, and wherein the optional booster dose is half of the initial dose.
136. The method of either of claims 134 or 135, wherein the optional booster dose is administered between about 90 minutes to about 120 minutes after the initial dose.
137. The method of claim 132, wherein the third, seventh, and eleventh sessions are MDMA-assisted psychotherapy sessions.
138. The method of any of claims 129-137, wherein the MDMA-assisted psychotherapy sessions last from between about 360 minutes to about 480 minutes.
139. The method of any of claims 129-138, wherein the evaluation of results occurs at 3 months, 6 months, and 9 months after a subject successfully completes the treatment protocol.
140. The method of any of claims 129-139, wherein, prior to conducting a plurality of psychotherapy sessions, the subject completes a group therapy preparation course.
141. The method of claim 140, wherein the group therapy preparation course includes a plurality of sessions.
142. The method of claim 141, wherein each of the plurality of sessions last for about 60 minutes.
143. The method of claim 142, wherein each of the plurality of sessions includes a discussion surrounding a specific topic.
144. The method of any of claims 141-143, wherein the plurality of sessions includes a first session, a second session, a third session, and a fourth session.
145. The method of claim 144, wherein the specific topic of the first session is building confidence; the specific topic of the second session is resources, barriers, and goal setting; the specific topic of the third session is increasing rewarding activities and building support; the specific topic of the fourth session is building the foundations for recovery.
146. The method of any of claims 129-146, wherein, prior to conducting a plurality of psychotherapy sessions, the subject completes a detoxification course.
147. A method of verifiably reducing alcohol consumption by a subject diagnosed with alcohol use disorder (AUD), comprising the steps of:
a. determining the amount of alcohol consumed by the subject prior to completing the method;
b. conducting a treatment protocol comprising 14 psychotherapy sessions, wherein 3 of the 14 psychotherapy sessions are MDMA-assisted psychotherapy sessions, and each of the MDMA-assisted psychotherapy sessions includes an initial dose, and optionally a booster dose, wherein the initial dose is between about 100 mg to about 150 mg, and the optional booster dose is about half of the initial dose;
c. measuring the amount of alcohol consumed of the subject three months, six months, and nine months after the final psychotherapy session;
d. verifying that the amount of alcohol consumed by the subject nine months after the final psychotherapy session is less than the amount consumed by the subject as determined prior to completing the method.
148. The method of claim 147, wherein the amount of alcohol consumed by the subject nine months after the final psychotherapy session is at least 50% less than the amount consumed by the subject as determined prior to completing the method.
149. The method of either of claims 148 or 149, wherein the amount of alcohol consumed by the subject nine months after the final psychotherapy session is at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% less than the amount consumed by the subject as determined prior to compl eting the method
150. A method of decreasing the daily alcohol consumption in a subject diagnosed with alcohol use disorder (AUD), the method comprising:
a. completing an 11 week psychotherapy regimen comprising 14 psychotherapy sessions, wherein 3 of the 14 psychotherapy sessions are about 360 minute to about 480 minute long MDMA-assisted psychotherapy sessions comprising the administration of two doses of MDMA, the two doses including an initial dose and a booster dose, the initial dose comprising between about 100 mg to about 150 mg of MDMA, and the booster dose, administered between about 90 minutes to about 120 minutes after the initial dose, comprising about half of the initial dose, and wherein 11 of the 14 psychotherapy sessions are non-drug psychotherapy sessions lasting about 60 minutes.
151. The method of claim 150, wherein the MDMA-assisted psychotherapy sessions occur once every 3 weeks for 9 weeks of the psychotherapy regimen, and the non-drug psychotherapy sessions occur once every week for all 11 weeks of the psychotherapy regimen, so that a non-dosing psychotherapy session follows an MDMA dosing session on weeks where the MDMA dosing session occurs.
152. A method of treating alcohol use disorder (A(JD) in a subject diagnosed therewith comprising a psychotherapy treatment protocol comprising the steps of:
a. conducting a screening visit, wherein the screening visit confirms the subject meets all inclusion criteria and does not meet any exclusion criteria;
b. completion of a plurality of psychotherapy sessions as part of a treatment protocol conducted under the supervision of a dyadic therapy team, the plurality of psychotherapy sessions including:
i. a first session, wherein the dyadic therapy pair introduces the therapy program to the subject, prepares the subject for the treatment protocol, and discusses the subject's personal story;
ii. a second session, wherein the dyadic therapy team teaches and practices a relaxation technique with the subject and reviews the subject's current recovery plan;
iii. a third session, wherein the subject is administered at least one dose of MDMA, wherein the at least one dose of MDMA is between about 100 mg to about 150 mg;
iv. a fourth session, wherein the dyadic therapist team guides the subject through a mindfulness routine, and the subject optionally provides a narrative of their experience after being administered at least one dose of MDMA;
v. a fifth session, wherein the dyadic therapy team discusses high risk situations that may lead to a relapse, and the workability of alcohol as a control strategy, with the subject;
vi. a sixth session, wherein the dyadic therapist team practices at least one defusion technique with the subject;
vii. a seventh session, wherein the subject is administered at least one dose of MDMA, wherein the at least one dose of MDMA is between about 100 mg to about 150 mg;
viii. an eighth session, wherein the dyadic therapist team guides the subject through a mindfulness routine, and the subject optionally provides a narrative of their experience after being administered at least one dose of MDMA;

ix. a ninth session, wherein the dyadic therapist team aids the subject in identifying problematic avoidance of internal experiences and practices at least one acceptance technique with the subject;
x. a tenth session, wherein the dyadic therapist team aids the subject in identifying at least one thing important in their life, at least one skill they possess, and at least one barrier that may impact their success;
xi. an eleventh session, wherein the subject is administered at least one dose of MDMA, wherein the at least one dose of MDMA is between about 100 mg to about 150 mg;
xii. a twelfth session, wherein the dyadic therapist team guides the subject through a mindfulness routine, and the subject optionally provides a narrative of the MDMA administration;
xiii. a thirteenth session, wherein the dyadic therapist team aids the subject in developing self-compassion, recognizing urges to consume alcohol, and practicing methods of establishing everyday mindfulness; and xiv. a fourteenth session, wherein the dyadic therapist team reviews the treatment protocol with the subject, and establishes an ongoing recovery plan with the subject.
153. The method of claim 152, wherein the at least one dose of MDMA comprises an initial dose, and optionally a booster dose, wherein the initial dose of MDMA
is between about 100 mg to about 150 mg, and the optional booster dose of MDMA is about half of the initial dose of MDMA.
154. MDMA for use in a method of treating alcohol use disorder (AUD) in a subject diagnosed therewith, comprising a psychotherapy treatment protocol comprising the steps of:
a. conducting a screening visit, wherein the screening visit confirms the subject meets all inclusion criteria and does not meet at least one exclusion criteria;
b. completion of a plurality of psychotherapy sessions as part of a treatment protocol conducted under the supervision of a dyadic therapy team, the plurality of psychotherapy sessions including:
i. a first session, wherein the dyadic therapy pair introduces the therapy program to the subject, prepares the subject for the treatment protocol, and discusses the subject's personal story;
ii. a second session, wherein the dyadic therapy team teaches and practices a relaxation technique with the subject and reviews the subject's current recovery plan;

iii. a third session, wherein the subject is administered at least one dose of MDMA, wherein the at least one dose of MDMA is between about 100 mg to about 150 mg;
iv. a fourth session, wherein the dyadic therapist team guides the subject through a mindfulness routine, and the subject optionally provides a narrative of their experience after being administered at least one dose of MDMA;
v. a fifth session, wherein the dyadic therapy team discusses high risk situations that may lead to a relapse, and the workability of alcohol as a control strategy, with the subject;
vi. a sixth session, wherein the dyadic therapist team practices at least one defusion technique with the subject;
vii. a seventh session, wherein the subject is administered at least one dose of MDMA, wherein the at least one dose of MDMA is between about 100 mg to about 150 mg, viii. an eighth session, wherein the dyadic therapist team guides the subject through a mindfulness routine, and the subject optionally provides a narrative of their experience after being administered at least one dose of MDMA;
ix. a ninth session, wherein the dyadic therapist team aids the subject in identifying problematic avoidance of internal experiences and practices at least one acceptance technique with the subject;
x. a tenth session, wherein the dyadic therapist team aids the subject in identifying at least one thing important in their life, at least one skill they possess, and at least one barrier that may impact their success, xi. an eleventh session, wherein the subject is administered at least one dose of MDMA, wherein the at least one dose of MDMA is between about 100 mg to about 150 mg, xii. a twelfth session, wherein the dyadic therapist team guides the subject through a mindfulness routine, and the subject optionally provides a naliative of the MDMA administiation, xiii. a thirteenth session, wherein the dyadic therapist team aids the subject in developing self-compassion, recognizing urges to consume alcohol, and practicing methods of establishing everyday mindfulness;
xiv. and a fourteenth session, wherein the dyadic therapist team reviews the treatment protocol with the subject, and establishes an ongoing recovery plan with the subject.
155. The method of claim 154, wherein the at least one dose of MDMA comprises an initial dose, and optionally a booster dose, wherein the initial dose of MDMA
is between about 100 mg to about 150 mg, and the optional booster dose of MDMA is about half of the initial dose of MDMA.
156. A method of reducing the units of alcohol consumed per week in a subject diagnosed with alcohol use disorder (AUD) by between about 70% to about 100% after completing the method, the method comprising:
a. determining the amount of alcohol consumed by the subject in units per week prior to completing the method;
b. conducting a treatment protocol comprising 14 psychotherapy sessions, wherein 3 of the 14 psychotherapy sessions are MDMA-assisted psychotherapy sessions, and each of the MDMA-assisted psychotherapy sessions includes an initial dose, and optionally a booster dose, wherein the initial dose is between about 100 mg to about 150 mg, and the optional booster dose is about half of the initial dose;
c. measuring the amount of alcohol consumed of the subject in units per week three months, six months, and nine months after completing the 14 psychotherapy sessions;
d. verifying that the amount of alcohol consumed by the subject nine months after completing the 14 psychotherapy sessions is between about 70% to about 100% less than the amount consumed by the subject as determined prior to completing the method.
157 The method of any of claims 129-156, wherein the subject has harmful use AUD.
158. The method of any of claims 129-156, wherein the subject has dependent use AUD.
159. The method of any of claims 129-156, wherein the subject has alcohol abuse, alcohol dependence, alcohol addiction, or alcoholism.
160. The method of any of claims 129-159, wherein the subject completes an alcohol detoxification course.
161. The method of claim 160, wherein the subject completes a pre-detoxification motivational group therapy course prior to the alcohol detoxification course.
162. The method of any of claims 129-159, wherein the subject completes a group therapy preparation course.
163. The method of any of claims 1-162, wherein the subject has a genetic variation associated with a substance use disorder, such as alcohol use disorder, and including a genetic variation in mCi1uR5 or or FKBP5.
164. The method of any of claims 1-163, wherein the subject is administered one or more therapeutic agents in addition to MDMA.
165. The method of claim 164, wherein the subject is also administered a therapeutically effective amount of an additional active agent selected from the group consisting of:
an opioid antagonist (e.g., nalmefene, naltrexone), a CB1 antagonist (e.g., rimonabant), a CRH1 receptor antagonist (e.g., verucerfont, pexacerfont), a antagonist (e.g., tradipitant), an OTR agonist (e.g., oxytocin), a GABA agent (e.g., topiramate, baclofen, a benzodiazepine, such as alprazolam, diazepam or lorazepam), a voltage-gated sodium channel inhibitor (e.g., oxacarbazepine, valproic acid, zonisamide), a voltage-dependent calcium channel agonist (e.g., gabapentin, pregabalin), an ct7 nicotinic acetylcholine receptor agonist (e.g., varenicline), a 5-HT3 antagonist (e.g., ondansetron), a 5-HT1A, receptor partial agonist (e.g., aripiprazole), a 5-HT2A receptor antagonist (e.g., quetiapine, olanzapine, mirtazapine), a 5-HT
reuptake inhibitor (e.g., trazodone), a SERT inhibitor (e.g., duloxetine), a al adrenoreceptor antagonist (e.g., doxazosin, prazosin), a glucocorticoid receptor antagonist (e.g., mifepri stone), an al adrenoreceptor agonist (e.g., guanfacine), an AChE inhibitor (e.g., citicoline), a dopamine D2 receptor antagonist (e.g., tiapride), an a2 adrenoreceptor agonist (e.g., clonidine), an NMDA receptor antagonist (e.g., acamprosate) and an aldehyde dehydrogenase inhibitor (e.g., disulfiram), and pharmaceutically acceptable salts thereof.
166. A pharmaceutical composition of MDMA for use in the method of any of claims 1-165, wherein said composition comprises a substantially pure MDMA compound haying a chromatogiaphic purity of greatet than 95% as measuied by HPLC.
167. The substantially pure MDMA of claim 131, wherein said substantially pure MDMA
is substantially free of impurities or adulterants.
168. A pharmaceutical composition of MDMA for use in the method of any of claims 1-130 wherein said MDMA is a pure or substantially pure individual enantiomer of R-MDMA or S-MDMA having an enantiomeric excess greater than 90%, or an enantiomerically enriched mixture having an enantiomeric excess in any amount greater than 0%.
169. A pharmaceutical composition of MDMA for use in the method of any of claims 1-165, wherein said MDMA is in unit dosage form.
170. The unit dosage form of claim 169, wherein said unit dosage form is an immediate release, controlled release, sustained release, extended release, or modified release formulation.
171. MDMA for use in a method of reducing the units of alcohol consumed per week in a subject diagnosed with alcohol use disorder (AUD) by between about 70% to about 100% after completing the method, the method comprising:

a. determining the amount of alcohol consumed by the subject in units per week prior to completing the method;
b. completion by the subject of a preparation group course;
c. conducting a treatment protocol comprising 14 psychotherapy sessions, wherein 3 of the 14 psychotherapy sessions are MDMA-assisted psychotherapy sessions, and each of the MDMA-assisted psychotherapy sessions includes an initial dose, and optionally a booster dose, wherein the initial dose is between about 100 mg to about 150 mg, and the optional booster dose is about half of the initial dose;
d. measuring the amount of alcohol consumed of the subject in units per week three months, six months, and nine months after completing the 14 psychotherapy sessions;
e. verifying that the amount of alcohol consumed by the subject nine months after completing the 14 psychotherapy sessions is between about 70% to about 100% less than the amount consumed by the subject as determined prior to completing the method.
172. MDMA for use in a method of decreasing the daily alcohol consumption in a subject diagnosed with alcohol use disorder, the method comprising: completing an 11 week psychotherapy regimen comprising 14 psychotherapy sessions, wherein 3 of the psychotherapy sessions are about 360 minute to about 480 minute long MDMA-assisted psychotherapy sessions comprising thc administration of two doses of MDMA, the two doses including an initial dose and a booster dose, the initial dose comprising between about 100 mg to about 150 mg of MDMA, and the booster dose, administered between about 90 minutes to about 120 minutes after the initial dose, comprising about half of the initial dose, and wherein 11 of the 14 psychotherapy sessions are non-drug psychotherapy sessions lasting about 60 minutes.
173. Use of MDMA for the manufacture of a medicament for a treatment for an alcohol use disorder (AUD) subject, according to the method of any of claims 1-165.
174. The use of claim 173, wherein the AUD subject has dependent use AUD, harmful use AUD, alcohol abuse, alcohol dependence, alcohol addiction, or alcoholism.
175. Use of MDMA in conjunction with psychotherapy to treat an alcohol use disorder (AUD) subject, according to the method of any of claims 1-165.
176. Use of MDMA for the manufacture of a medicament for a treatment to reduce alcohol use by an AUD subject, according to the method of any of claims 1-165.
177. Use of MDMA for the manufacture of a medicament for a treatment to prevent relapse into alcohol use by an AUD subject, according to the method of any of claims 1-165.
178. Use of MDMA for the manufacture of a medicament for a treatment to promote alcohol abstinence by an AUD subject, according to the method of any of claims 165.
179. Use of MDMA for the manufacture of a medicament for the treatment of the symptoms of depression or anxiety associated with AUD, according to the method of any of claims 1-165.
180. Use of MDMA for the manufacture of a medicament for the treatment of the symptoms of a psychiatric disorder comorbid with AUD, according to the method of any of claims 1-165.
181. The use of claim 180, wherein the psychiatric disorder comorbid with AUD
is any of post-traumatic stress disorder (PTSD), adjustment disorder, affective disorder, depression, atypical depression, postpartum depression, catatonic depression, a depressive disorder due to a medical condition, premenstrual dysphoric disorder, seasonal affective disorder, dysthymia, anxiety, phobia disorders, binge disorders, body dysmorphic disorder, a mood disorder related to another health condition, disruptive behavior disorders, eating disorders, impulse control disorders, obsessive compulsive disorder (OCD), attention deficit hyperactivity disorder (ADRD), personality disorders, attachment disorders, dissociative disorders, or substance-related disordeis including substance use disolder (SUD).
182. The use of claim 180, wherein the psychiatric disorder comorbid with AUD
is a behavioral addiction, including any of pathological gambling, kleptomania, pyromania, compulsive buying, compulsive sexual behaviour, Internet addiction, pornography, and binge eating disorder.
183. The use of any of claims 173-182, wherein AUD is diagnosed as defined by a version of the Diagnostic and Statistical Manual of Mental Disorders (DSM), wherein diagnosis comprises scoring above the diagnostic threshold for AUD.
184. The use of any of claims 173-183, wherein severity of AUD is determined using one or more questionnaires.
185. The use of claim 184, wherein the one or more questionnaire(s) comprise the Severity of Alcohol Questionnaire (SADQ) and/or the Short Inventory of Problems for Alcohol (SIP) questionnaire.
186. The use of either of claims 184 or 185, wherein the one or more questionnaire(s) comprise the Severity of Alcohol Questionnaire (SADQ).
187. rt he use of claim 186, wherein the severity of AUD is scored on the SADQ
as mild (score of less than 16), moderate (score of 16-30), or severe (score of greater than or equal to 31).
188. The use of claim 187, wherein the severity of AUD is mild (score of less than 16).
189. The use of claim 187, wherein the severity of AUD is moderate (score of 16-30).
190. The use of claim 187, wherein the severity of AUD is severe (score of greater than or equal to 31).
191. The use of any of claims 173-190, comprising monitoring adverse events during MDMA-assisted psychotherapy.
192. The use of claim 190, wherein the adverse events comprise one or more of hyperthermia, hypertension, and elevated heart rate.
193. The use of either of claims 191 or 192, wherein adverse events are not observed during MDMA-assisted psychotherapy.
194. The use of any of claims 173-193, comprising monitoring for adverse events after administration of MDMA-assisted psychotherapy.
195. The use of claim 194, wherein the adverse events after MDMA-assisted psychotherapy comprise mood disturbances and/or depressed affect.
196. The use of claim 195, wherein a mood disturbance and/or depressed affect after administration of MDMA-assisted psychotherapy are not observed and/or reported.
197. The use of claim 196, wherein mood disturbances and/or depressed affect are not observed and/or reported within about one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, or ten days after MDMA-assisted psychotherapy.
198. The use of any of claims 194-197, wherein adverse events after administration of MDMA-assisted psychotherapy are not observed or reported.
199. The use of any of claims 173-198, comprising monitoring for adverse events both during and after administration of MDMA.
200. The use of claim 199, wherein the adverse events during administration of MDMA
comprise one or more of hyperthermia, hypertension, and elevated heart rate, and the adverse events after administration of MDMA comprise mood disturbances and/or depressed affect.
201. The use of either of claims 199 or 200, wherein no adverse events are observed or reported during or after administration of MDMA-assisted psychotherapy.
202. The use of claim 201, wherein no adverse events are observed or reported during administration of MDMA, or reported after administration of MDMA, including within about one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, or ten days after administration.
203. The use of any of claims 173-202, wherein the subject has a genetic variation associated with a substance use disorder, such as alcohol use disorder, and including a genetic variation in mGluR5 or or FKBP5.
204. The use of any of claims 173-203, wherein the subject is administered one or more therapeutic agents in addition to MDMA.
205. The use of claim 204, wherein the subject is also administered a therapeutically effective amount of an additional active agent selected from the group consisting of:
an opioid antagonist (e.g., nalmefene, naltrexone), a CB1 antagonist (e.g., rimonabant), a CRH1 receptor antagonist (e.g., verucerfont, pexacerfont), a antagonist (e.g., tradipitant), an OTR agonist (e.g., oxytocin), a GABA agent (e.g., topiramate, baclofen, a benzodiazepine, such as alprazolam, diazepam or lorazepam), a voltage-gated sodium channel inhibitor (e.g., oxacarbazepine, valproic acid, zonisamide), a voltage-dependent calcium channel agonist (e.g., gabapentin, pregabalin), an a7 nicotinic acetylcholine receptor agonist (e.g., varenicline), a 5-HT3 antagonist (e.g., ondansetron), a 5-HT1A, receptor partial agonist (e.g., aripiprazole), a 5-HT2A receptor antagonist (e.g., quetiapine, olanzapine, mirtazapine), a 5-HT
reuptake inhibitor (e.g., trazodone), a SERT inhibitor (e.g., duloxetine), a al adrenoreceptor antagonist (e.g., doxazosin, prazosin), a glucocorticoid receptor antagonist (e.g., mifepristone), an al adrenoreceptor agonist (e.g., guanfacine), an AChE inhibitor (e.g., citicoline), a dopamine D2 receptor antagonist (e.g., tiapride), an a2 adrenoreceptor agonist (e.g., cloni dine), an NMDA receptor antagonist (e.g., acamprosate) and an aldehyde dehydrogenase inhibitor (e.g., disulfiram), and pharmaceutically acceptable salts thereof.
CA3204457A 2021-01-06 2022-01-06 Mdma in the treatment of alcohol use disorder Pending CA3204457A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US202163134582P 2021-01-06 2021-01-06
US63/134,582 2021-01-06
US202163137615P 2021-01-14 2021-01-14
US63/137,615 2021-01-14
PCT/US2022/011511 WO2022150525A1 (en) 2021-01-06 2022-01-06 Mdma in the treatment of alcohol use disorder

Publications (1)

Publication Number Publication Date
CA3204457A1 true CA3204457A1 (en) 2022-07-14

Family

ID=80050571

Family Applications (1)

Application Number Title Priority Date Filing Date
CA3204457A Pending CA3204457A1 (en) 2021-01-06 2022-01-06 Mdma in the treatment of alcohol use disorder

Country Status (2)

Country Link
CA (1) CA3204457A1 (en)
WO (1) WO2022150525A1 (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3221280A1 (en) 2021-06-03 2022-12-08 Arcadia Medicine, Inc. Enantiomeric entactogen compositions and methods of their use
WO2023019369A1 (en) * 2021-08-20 2023-02-23 Pharmala Biotech Inc. Compositions comprising non-racemic mixtures of (r)- and (s)-3,4-methylenedioxymethamphetamine or (r) and (s) n-methyl-1,3-benzodioxolylbutanamine and uses thereof
US11845736B2 (en) 2021-10-01 2023-12-19 Empathbio, Inc. Prodrugs of MDMA, MDA, and derivatives thereof
WO2023057953A1 (en) * 2021-10-07 2023-04-13 Clearmind Medicine Inc. 5-mapb or 6-apb for use in drug-assisted psychotherapy
WO2023129958A2 (en) 2021-12-28 2023-07-06 ATAI Life Sciences AG Nitric oxide releasing prodrugs of mda and mdma
US20230218568A1 (en) * 2022-01-07 2023-07-13 Universitatsspital Basel R-MDMA and S-MDMA TO ASSIST PSYCHOTHERAPY
US20240082212A1 (en) * 2022-09-09 2024-03-14 ATAI Life Sciences AG Use of mdma for treatment of stress-related disorders
WO2024064825A1 (en) * 2022-09-21 2024-03-28 Axsome Therapeutics, Inc. Compounds and combinations thereof for treating neurological and psychiatric conditions

Family Cites Families (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE279194C (en)
DE274350C (en) 1912-12-24 1914-05-16 Merck E
US4327725A (en) 1980-11-25 1982-05-04 Alza Corporation Osmotic device with hydrogel driving member
US4624848A (en) 1984-05-10 1986-11-25 Ciba-Geigy Corporation Active agent containing hydrogel devices wherein the active agent concentration profile contains a sigmoidal concentration gradient for improved constant release, their manufacture and use
GB8518301D0 (en) 1985-07-19 1985-08-29 Fujisawa Pharmaceutical Co Hydrodynamically explosive systems
EP0230654B1 (en) 1985-12-28 1992-03-18 Sumitomo Pharmaceuticals Company, Limited Sustained pulsewise release pharmaceutical preparation
US4968509A (en) 1987-07-27 1990-11-06 Mcneilab, Inc. Oral sustained release acetaminophen formulation and process
IL92966A (en) 1989-01-12 1995-07-31 Pfizer Dispensing devices powered by hydrogel
US5017381A (en) 1990-05-02 1991-05-21 Alza Corporation Multi-unit pulsatile delivery system
KR0182801B1 (en) 1991-04-16 1999-05-01 아만 히데아키 Method of manufacturing solid dispersion
US5229135A (en) 1991-11-22 1993-07-20 Prographarm Laboratories Sustained release diltiazem formulation
WO1993009785A1 (en) 1991-11-22 1993-05-27 Procter & Gamble Pharmaceuticals, Inc. Risedronate delayed-release compositions
US5461140A (en) 1992-04-30 1995-10-24 Pharmaceutical Delivery Systems Bioerodible polymers for solid controlled release pharmaceutical compositions
US5260068A (en) 1992-05-04 1993-11-09 Anda Sr Pharmaceuticals Inc. Multiparticulate pulsatile drug delivery system
CA2147283C (en) 1992-10-16 2007-01-16 Kouichi Nakamichi Method of manufacturing wax matrices
US5260069A (en) 1992-11-27 1993-11-09 Anda Sr Pharmaceuticals Inc. Pulsatile particles drug delivery system
US5686105A (en) 1993-10-19 1997-11-11 The Procter & Gamble Company Pharmaceutical dosage form with multiple enteric polymer coatings for colonic delivery
US5567441A (en) 1995-03-24 1996-10-22 Andrx Pharmaceuticals Inc. Diltiazem controlled release formulation
CA2220451A1 (en) 1995-05-17 1996-11-21 Cedars-Sinai Medical Center Methods and compositions for improving digestion and absorption in the small intestine
US5837284A (en) 1995-12-04 1998-11-17 Mehta; Atul M. Delivery of multiple doses of medications
US5840329A (en) 1997-05-15 1998-11-24 Bioadvances Llc Pulsatile drug delivery system
US6395300B1 (en) 1999-05-27 2002-05-28 Acusphere, Inc. Porous drug matrices and methods of manufacture thereof
US6465014B1 (en) 2001-03-21 2002-10-15 Isp Investments Inc. pH-dependent sustained release, drug-delivery composition
JP7273037B2 (en) 2017-08-03 2023-05-12 マインド メディシン, インコーポレイテッド Use of 3-methylmethcathinone

Also Published As

Publication number Publication date
WO2022150525A9 (en) 2023-04-20
WO2022150525A1 (en) 2022-07-14

Similar Documents

Publication Publication Date Title
CA3204457A1 (en) Mdma in the treatment of alcohol use disorder
US20220169668A1 (en) Methods of treating neurocognitive disorders, chronic pain and reducing inflammation
Cullen et al. N-acetylcysteine for nonsuicidal self-injurious behavior in adolescents: an open-label pilot study
Arnold et al. Placebo-controlled pilot trial of mecamylamine for treatment of autism spectrum disorders
CA3072780C (en) 3-methyl methcathinone in psychotherapeutic intervention
Shahrokh et al. The language of mental health: A glossary of psychiatric terms
US20230091682A1 (en) Cognitive disorder prevention and therapy
US20240122895A1 (en) Entactogen-assisted therapy for alcohol use disorder and addiction-related conditions and comorbidities
EP4337316A1 (en) Therapeutic aminoindane compounds and compositions
Baggott et al. 3, 4-Methylenedioxymethamphetamine (MDMA): A review of the English-language scientific and medical literature
CN112789045A (en) Compositions and methods for treating autism
Aouad The psychological disturbances of the child undergoing Surgery-from admission till beyond discharge
US20230381168A1 (en) Adjunctive therapy for depression
CA3220228A1 (en) Ketamine in the treatment of behavioural addictions
WO2023172701A2 (en) Therapeutic combinations, compositions, and methods for designing and producing entactogenic mindstates
Zeise Clinical Neuropharmacology
WO2024057192A1 (en) Methods of treating fibromyalgia with compositions comprising psilocybin
WO2024054452A1 (en) Methods for the treatment of mild cognitive impairment
WO2023086252A1 (en) Treatment of treatment resistant depression with psilocybin
Saklayen Effects of propranolol on cognition and eye contact in autism spectrum disorder (ASD)
Moore et al. Anxiety disorders in children and adolescents
Wong SPECIAL SYMPOSIUM
Owens Case 2.16 Treating Anxiety
Owens Treating Anxiety