WO2023230476A1 - Methods and compositions for preventing or delaying type 1 diabetes - Google Patents

Methods and compositions for preventing or delaying type 1 diabetes Download PDF

Info

Publication number
WO2023230476A1
WO2023230476A1 PCT/US2023/067357 US2023067357W WO2023230476A1 WO 2023230476 A1 WO2023230476 A1 WO 2023230476A1 US 2023067357 W US2023067357 W US 2023067357W WO 2023230476 A1 WO2023230476 A1 WO 2023230476A1
Authority
WO
WIPO (PCT)
Prior art keywords
day
subject
antibody
diabetes
less
Prior art date
Application number
PCT/US2023/067357
Other languages
French (fr)
Inventor
Francisco Leon
Eleanor L. RAMOS
Original Assignee
Provention Bio, Inc.
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 Provention Bio, Inc. filed Critical Provention Bio, Inc.
Priority claimed from US18/321,964 external-priority patent/US20230382993A1/en
Priority claimed from TW112119143A external-priority patent/TW202411250A/en
Publication of WO2023230476A1 publication Critical patent/WO2023230476A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • the present disclosure relates in general to compositions and methods of preventing or delaying the onset of clinical type 1 diabetes (T1D) in subjects at risk, and more particularly the use of anti-CD3 antibodies.
  • T1D clinical type 1 diabetes
  • Type 1 diabetes is caused by the autoimmune destruction of insulin producing beta cells in the islets of Langerhans leading to dependence on exogeneous insulin injections for survival.
  • T1D Type 1 diabetes
  • Most affected individuals with T1D are not able to consistently achieve desired glycemic targets.
  • For individuals with type 1 diabetes there are persisting concerns for increased risk of both morbidity and mortality. Two recent studies noted loss of 17.7 life-years for children diagnosed before age 10, and 11 and 13 life-years lost for adult-diagnosed Scottish men and women respectively.
  • T1D progresses through asymptomatic stages prior to overt hyperglycemia, characterized first by the appearance of autoantibodies (Stage 1) and then dysglycemia (Stage 2).
  • Stage 2 metabolic responses to a glucose load are impaired but other metabolic indices, for example glycosylated hemoglobin, are normal and insulin treatment is not needed.
  • FcR non-binding anti-CD3 monoclonal antibody teplizumab One promising therapy is the FcR non-binding anti-CD3 monoclonal antibody teplizumab, as several studies have shown that short-term treatment reduces loss of P cell function durably, with an observable effect seen as long as 7 years after diagnosis and treatment.
  • the drug modifies the function of CD8+ T lymphocytes, which are thought to be important effector cells that cause beta cell killing.
  • aspects of the disclosure relate to a method of preventing or delaying onset of clinical type 1 diabetes (T1D), comprising: administering a prophylactically effective amount of an anti- CD3 antibody to a non-diabetic subject who is at risk for T1D.
  • T1D clinical type 1 diabetes
  • the method of preventing or delaying onset of clinical type 1 diabetes comprises administering a prophylactically effective amount of an anti-CD3 antibody to a non-diabetic subject who is at risk of T1D, wherein the prophylactically effective amount has a cumulative dose of from about 10,500 pg/m 2 to about 14,000 pg/m 2 .
  • the non-diabetic subject is an adult or a pediatric subject.
  • the pediatric subject is 8 years old or older. In some embodiments, the pediatric subject is 7 years old or older. In some embodiments, the pediatric subject is 6 years old or older. In some embodiments, the pediatric subject is 5 years old or older. In some embodiments, the pediatric subject is 4 years old or older. In some embodiments, the pediatric subject is 3 years old or older. In some embodiments, the pediatric subject is 3 years old or older. In some embodiments, the pediatric subject is 2 year old or older. In some embodiments, the pediatric subject is 1 year old or older. In some embodiments, the pediatric subject is 1 year old or younger. In some embodiments, the pediatric subject is an infant.
  • the non-diabetic subject is a relative of a patient with T1D.
  • the method further includes determining that the non-diabetic subject (1) is negative for zinc transporter 8 (ZnT8) antibodies, (2) is HLA-DR4+, and/or (3) is not HLA-DR3+.
  • ZnT8 zinc transporter 8
  • the non-diabetic subject has 2 or more diabetes-related autoantibodies selected from islet cell antibodies (ICA), insulin autoantibodies (IAA), and antibodies to glutamic acid decarboxylase (GAD), ty rosine phosphatase (IA-2/ICA512) or ZnT8.
  • ICA islet cell antibodies
  • IAA insulin autoantibodies
  • GAD glutamic acid decarboxylase
  • IA-2/ICA512 ty rosine phosphatase
  • ZnT8 ZnT8.
  • the non-diabetic subject has abnormal glucose tolerance on oral glucose tolerance test (OGTT).
  • OGTT oral glucose tolerance test
  • the abnormal glucose tolerance on OGTT is a fasting glucose level of 110-125 mg/dL, or 2 hour plasma of > 140 and ⁇ 200 mg/dL, or an intervening glucose value at 30, 60, or 90 minutes on OGTT > 200 mg/dL.
  • CGM glucose monitoring system
  • the non-diabetic subject does not have antibodies against ZnT8.
  • the non-diabetic subject is HLA-DR4+ and is not HLA-DR3+.
  • the non-diabetic subject does not have antibodies against ZnT8, is HLA-DR4+ and is not HLA-DR3+.
  • the non-diabetic subject is a relative of a patient with T1D and does not have antibodies against ZnT8. In some embodiments, the non-diabetic subject is a relative of a patient with T1D, does not have antibodies against ZnT8, is HLA-DR4+ and is not HLA-DR3+.
  • the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab. In some embodiments, the anti-CD3 antibody is teplizumab.
  • the prophylactically effective amount of the antibody comprises a 10 to 14-day course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at a cumulative dose greater than 10,500 micrograms/meter squared (pg/m 2 ). In some embodiments, the prophylactically effective amount of the antibody comprises a 14-day course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at a cumulative dose of from about 11,000 pg/m 2 to about 14,000 pg/m 2 . In some embodiments, the anti-CD3 antibody is or comprises teplizumab.
  • the prophylactically effective amount of the antibody comprises a 10 to 14 day course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 10,500 to about 14,000 pg/m 2 , from about
  • the prophylactically effective amount of the antibody comprises a 10 to 14-day course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at about 10,500 pg/m 2 , 11,000 pg/m 2 , 11,500 pg/m 2 , 12,000 pg/m 2 , 12,500 pg/m 2 , 13,000 pg/m 2 , 13,500 pg/m 2 , or 14,000 pg/m 2 .
  • the anti-CD3 antibody is or comprises teplizumab.
  • the method comprises administering to the non-diabetic subj ect who is at risk for T1D a 14-day course IV infusion of the anti-CD3 antibody at about 60 pg/m 2 , about 125 pg/m 2 , about 250 pg/m 2 , and about 500 pg/m 2 , on days 1-4, respectively, and a dose of about 1,000 pg/m 2 on each of days 5-14. In some embodiments, the cumulative dose is about 10,935 pg/m 2 .
  • the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab.
  • the anti-CD3 antibody is teplizumab.
  • the method comprises administering to the non-diabetic subject who is at risk for T1 D a 14-day course IV infusion of teplizumab at about 60 pg/m 2 , about 125 pg/m 2 , about 250 pg/m 2 , and about 500 pg/m 2 , on days 1-4, respectively, and a dose of about 1,000 pg/m 2 on each of days 5-14.
  • the method comprises administering to the non-diabetic subj ect who is at risk for T1D a 14-day course IV infusion of the anti-CD3 antibody at about 60 pg/m 2 , about 125 pg/m 2 , about 250 pg/m 2 , and about 500 pg/m 2 , on days 1-4, respectively, and a dose of about 1,030 pg/m 2 on each of days 5-14. In some embodiments, the cumulative dose is about 11,235 pg/m 2 .
  • the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab.
  • the anti-CD3 antibody is teplizumab.
  • the method comprises administering to the non-diabetic subject who is at risk for T1D a 14-day course IV infusion of teplizumab at about 60 pg/m 2 , about 125 pg/m 2 , about 250 pg/m 2 , and about 500 pg/m 2 , on days 1-4, respectively, and a dose of about 1,030 pg/m 2 on each of days 5-14.
  • the method comprises administering to the non-diabetic subj ect who is at risk for T1D a 14-day course IV infusion of the anti-CD3 antibody at about 100 pg/m 2 , about 425 pg/m 2 , about 850 pg/m 2 , and about 850 pg/m 2 , on days 1-4, respectively, and a dose of about 1,000 pg/m 2 on each of days 5-14. In some embodiments, the cumulative dose is about 12,225 pg/m 2 .
  • the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab.
  • the anti-CD3 antibody is teplizumab.
  • the method comprises administering to the non-diabetic subject who is at risk for T1D a 14-day course IV infusion of teplizumab at about 100 pg/m 2 , about 425 pg/m 2 , about 850 pg/m 2 , and about 850 pg/m 2 , on days 1-4, respectively, and a dose of about 1,000 pg/m 2 on each of days 5-14.
  • the method comprises administering to the non-diabetic subj ect who is at risk for T1D a 14-day course IV infusion of the anti-CD3 antibody at about 65 pg/m 2 , about 125 pg/m 2 , about 250 pg/m 2 , and about 500 pg/m 2 , on days 1-4, respectively, and a dose of about 1,070 pg/m 2 on each of days 5-14.
  • the cumulative dose is about 11,640 pg/m 2 .
  • the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab.
  • the anti-CD3 antibody is teplizumab.
  • the method comprises administering to the non-diabetic subject who is at risk for T1D a 14-day course IV infusion of teplizumab at about 65 pg/m 2 , about 125 pg/m 2 , about 250 pg/m 2 , and about 500 pg/m 2 , on days 1-4, respectively, and a dose of about 1,070 pg/m 2 on each of days 5-14.
  • the method comprises administering to the non-diabetic subj ect who is at risk for T1D a 14-day course IV infusion of the anti-CD3 antibody at about 65 pg/m 2 , about 125 pg/m 2 , about 250 pg/m 2 , and about 500 pg/m 2 , on days 1-4, respectively, and a dose of about 1,030 pg/m 2 on each of days 5-14.
  • the cumulative dose is about 11,240 pg/m 2 .
  • the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab.
  • the anti-CD3 antibody is teplizumab.
  • the method comprises administering to the non-diabetic subject who is at risk for T1D a 14-day course IV infusion of teplizumab at about 65 pg/m 2 , about 125 pg/m 2 , about 250 pg/m 2 , and about 500 pg/m 2 , on days 1-4, respectively, and a dose of about 1,030 pg/m 2 on each of days 5-14.
  • the prophylactically effective amount delays median time to clinical diagnosis of T1D by at least 50%, at least 80%, or at least 90%. In some embodiments, the prophylactically effective amount delays median time to clinical diagnosis of T1D by at least 12 months, at least 18 months, at least 24 months, at least 36 months, at least 48 months, or at least 60 months.
  • the method further comprises determining, prior to or after the administering step, that the non-diabetic subject has more than about 10% TIGIT+KLRG1+CD8+ T-cells in all CD3+ T cells, which is indicative of successful prevention or delay of the onset of clinical T1D.
  • the step of determining of TIGIT+KLRG1+CD8+ T-cells is by flow cytometry.
  • the method further comprises determining a decrease in a percentage of CD8+ T cells expressing proliferation markers Ki67 and/or CD57.
  • Some aspects of the disclosure relate to a method of preventing or delaying onset of clinical type 1 diabetes (T1D), comprising: administering to a non-diabetic subject 8 years of age or older who is at risk of T1D a 14 day course IV infusion of teplizumab at about 65 pg/m 2 on day 1, about 125 pg/m 2 on day 2, about 250 pg/m 2 on day 3, and about 500 pg/m 2 on day 4, and a dose of about 1,030 pg/m 2 on each of days 5-14.
  • T1D clinical type 1 diabetes
  • Some aspects of the disclosure relate to a method of delaying onset of Stage 3 type 1 diabetes (T1D), comprising administering to a subject in need thereof who has a diagnosis of stage 2 T1D a 14 day course IV infusion of teplizumab at about 65 pg/m 2 on day 1, about 125 pg/m 2 on day 2, about 250 pg/m 2 on day 3, and about 500 pg/m 2 on day 4, and a dose of about 1,030 pg/m 2 on each of days 5-14.
  • the non-diabetic subject is an adult or a pediatric subject.
  • the pediatric subject is 8 years old or older.
  • the pediatric subject is 7 years old or older.
  • the pediatric subject is 6 years old or older. In some embodiments, the pediatric subject is 5 years old or older. In some embodiments, the pediatric subject is 4 years old or older. In some embodiments, the pediatric subject is 3 years old or older. In some embodiments, the pediatric subject is 3 years old or older. In some embodiments, the pediatric subject is 2 year old or older. In some embodiments, the pediatric subject is 1 year old or older. In some embodiments, the pediatric subject is 1 year old or younger. In some embodiments, the pediatric subject is an infant. [0031] In some embodiments, the method comprises documenting at least two positive pancreatic islet autoantibodies in the subject who has dysglycemia without overt hyperglycemia before administering the 14 day course.
  • the subj ect in need thereof has dysglycemia without overt hyperglycemia and has two or more pancreatic islet autoantibodies.
  • the two or more pancreatic islet autoantibodies comprise islet cell antibodies (ICA), insulin autoantibodies (IAA), and antibodies to glutamic acid decarboxylase (GAD), tyrosine phosphatase (IA-2/ICA512) or ZnT8.
  • ICA islet cell antibodies
  • IAA insulin autoantibodies
  • GAD glutamic acid decarboxylase
  • IA-2/ICA512 tyrosine phosphatase
  • ZnT8 ZnT8.
  • the method comprises administering on at least each of days 1- 5, and prior to the administering of the 14 days course IV infusion, an effective amount of a nonsteroidal anti-inflammatory drug (NSAID), acetaminophen, an antihistamine, an antiemetic or a combination thereof.
  • NSAID nonsteroidal anti-inflammatory drug
  • the method comprises administering orally the NSAID, acetaminophen, antihistamine, antiemetic or combination thereof.
  • the method comprises administering the 14 days course IV infusion once daily for 14 consecutive days over a period of at least 30 minutes.
  • the method further comprises determining, pnor to or after the administering step, that the non-diabetic subject has more than about 10% TIGIT+KLRG1+CD8+ T-cells in all CD3+ T cells, which is indicative of successful prevention or delay of the onset of clinical T1D.
  • the determining of TIGIT+KLRG1+CD8+ T-cells is by flow cytometry.
  • the method further includes determining a decrease in a percentage of CD8+ T cells expressing proliferation markers Ki67 and/or CD57.
  • Other aspects of the disclosure relate to a method of prognosing responsiveness of an anti-CD3 antibody in preventing or delaying onset of type I diabetes (T1D), the method comprising administering a prophy tactically effective amount of an anti-CD3 antibody to a nondiabetic subject who is at risk for T1D, wherein the prophylactically effective amount has a cumulative dose of about 10,500 pg/rrr to about 14,000 pg/m 2 ; and determining C-peptide area under the curve (AUC): glucose AUC ratio, wherein an increase in the ratio indicates responsiveness to the anti-CD3 antibody and/or non-progression to clinical T1D.
  • AUC C-peptide area under the curve
  • an anti-CD3 antibody for use in a method of preventing or delaying onset of clinical type 1 diabetes (T1D), comprising administering a prophylactically effective amount of an anti-CD3 antibody to a non-diabetic subject who is at risk of T1D, wherein the prophylactically effective amount has a cumulative dose of about 10,500 pg/m 2 to about 14,000 pg/m 2 .
  • the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab.
  • the anti-CD3 antibody is teplizumab.
  • aspects of the disclosure relate to a method of preventing or delaying onset of Stage 3 type 1 diabetes (T1D), comprising administering to a subject who has a diagnosis of stage 2 T1D a 14 day course IV infusion of teplizumab at about 65 pg/m 2 on day 1, about 125 pg/m 2 on day 2, about 250 pg/m 2 on day 3, and about 500 pg/m 2 on day 4, and a dose of about 1,030 pg/m 2 on each of days 5-14.
  • T1D Stage 3 type 1 diabetes
  • the subject in need thereof has two or more pancreatic islet autoantibodies.
  • the subject in need thereof has dysglycemia without overt hyperglycemia and has two or more pancreatic islet autoantibodies.
  • the method comprises confirming stage 2 T1D by documenting at least two positive pancreatic islet autoantibodies in subjects who have dysglycemia without overt hyperglycemia before administering the 14 day course.
  • pancreatic islet autoantibodies comprise islet cell antibodies (ICA), insulin autoantibodies (IAA), antibodies to glutamic acid decarboxylase (GAD), tyrosine phosphatase (IA-2/ICA512) or ZnT8.
  • ICA islet cell antibodies
  • IAA insulin autoantibodies
  • GAD glutamic acid decarboxylase
  • IA-2/ICA512 tyrosine phosphatase
  • ZnT8 ZnT8.
  • FIG. 1 shows AUCinf, Cmax and Ctroughis: Reference Regimen and Simulated Alternative Regimens (Regimen A, Regimen B, regimen C). Exposures for the reference regimen reflect the Herold 14-day regimen administered to PROTECT study participants who received the anti- CD3 antibody. Exposures for alternative regimens reflect simulated anti-CD3 antibody administered to a similar population of T1D patients as a subset of PROTECT study participants.
  • the boxes indicate the median (line within a box) values as well as QI to Q3 interquartile ranges (IQRs).
  • the whiskers indicate Ql-1.5 IQR and Q3+1.5 IQR. The data points outside the whiskers indicate outlier values.
  • FIG. 2 shows AUCinf, Cmax and Ctroughis: Historical Clinical Trials and Simulated Alternative Regimens for the anti-CD3 antibody.
  • Protege and TN-I0 exposures represent all patients who received the Herold 14-day regimen in these studies;
  • PROTECT exposures represent patients who received the anti-CD3 antibody in the PROTECT study.
  • Regimen A, Regimen B and Regimen C graphs are simulated PK data for anti-CD3 antibody Regimen A, Regimen B and Regimen C using in a similar population of T1D patients as a subset of PROTECT study subjects.
  • the boxes indicate the median (line within a box) values as well as QI to Q3 interquartile ranges (IQRs).
  • the whiskers indicate Ql-1.5 IQR and Q3+1.5 IQR. The data points outside the whiskers indicate outlier values.
  • FIG. 3 shows Predicted Cmax and Ctrough Levels During 14-day Treatment Course: Regimens A and B.
  • the boxes indicate the median (line within a box) values as well as QI to Q3 interquartile ranges (IQRs).
  • the whiskers indicate Ql-1.5 IQR and Q3+1.5 IQR.
  • the data points outside the whiskers indicate outlier values.
  • FIG. 4 shows Predicted Cmax and Ctrough Levels During 14-day Treatment Course: Regimen C.
  • the boxes indicate the median (line within a box) values as well as QI to Q3 interquartile ranges (IQRs).
  • the whiskers indicate Ql-1.5 IQR and Q3+1.5 IQR.
  • the data points outside the whiskers indicate outlier values.
  • FIG. 5 shows AUCinf, Cmax and Ctroughis: Reference Regimen and Simulated Regimen D. Exposures for the reference regimen reflect the Herold 14-day regimen administered to PROTECT study participants who received the anti-CD3 antibody. Exposures for alternative regimens reflect simulated anti-CD3 antibody administered to a similar population of T1D patients as a subset of PROTECT study participants.
  • the boxes indicate the median (line within a box) values as well as QI to Q3 interquartile ranges (IQRs).
  • the whiskers indicate Ql-1.5 IQR and Q3+1.5 IQR. The data points outside the whiskers indicate outlier values.
  • FIG. 6 shows AUCinf, Cmax and Ctmughis: Historical Clinical Trials and Simulated Regimen D for the anti-CD3 antibody.
  • Protege and TN-10 exposures represent all patients who received the Herold 14-day regimen in these studies;
  • PROTECT exposures represent patients who received the anti-CD3 antibody in the PROTECT study.
  • Regimen D is simulated PK data for anti-CD3 antibody Regimen D using in a similar population of T1D patients as a subset of PROTECT study subjects.
  • the boxes indicate the median (line within a box) values as well as QI to Q3 interquartile ranges (IQRs).
  • the whiskers indicate Ql-1.5 IQR and Q3+1.5 IQR. The data points outside the whiskers indicate outlier values.
  • FIG. 7 shows Predicted Cmax and Ctrough Levels During 14-day Treatment Course: Regimen D.
  • the boxes indicate the median (line within a box) values as well as QI to Q3 interquartile ranges (IQRs).
  • the whiskers indicate Ql-1.5 IQR and Q3+1.5 IQR.
  • the data points outside the whiskers indicate outlier values.
  • a method of preventing or delaying the onset of clinical type 1 diabetes comprising: providing a non-diabetic subject who is at risk for T1D; administering a prophylactically effective amount of an anti-CD3 antibody to the nondiabetic subject; and determining, prior to or after the administering step, that the non-diabetic subject has more than about 10% TIGIT+KLRG1+CD8+ T-cells in all CD3+ T cells, which is indicative of successful prevention or delay of the onset of clinical T1D.
  • a method of prognosing responsiveness of an anti-CD3 antibody in preventing or delaying the onset of T1D.
  • the method can include: providing a non-diabetic subject who is at risk for T1D; administering a prophylactically effective amount of the anti-CD3 antibody, e.g., tephzumab, to the non-diabetic subject; and determining C-peptide area under the curve (AUC): glucose AUC ratio, wherein an increase in the ratio indicates responsiveness to the anti-CD3 antibody.
  • AUC C-peptide area under the curve
  • the articles “a” and “an” refer to one or more than one, e.g., to at least one, of the grammatical object of the article.
  • the use of the words “a” or “an” when used in conjunction with the term “comprising” herein may mean “one,” but it is also consistent with the meaning of "one or more,” “at least one,” and “one or more than one.”
  • “about” and “approximately” generally mean an acceptable degree of error for the quantity measured given the nature or precision of the measurements. Exemplary degrees of error are within 30 percent (%), within 25% within 20 %, for example, within 10%, or, within 5% of a given range of values. The term “substantially” means more than 50%, more than 60%, more than 70% more than 80%, and more than 90% or 95%.
  • “comprising” or “comprises” is used in reference to compositions, methods, and respective component(s) thereof, that are present in a given embodiment, yet open to the inclusion of unspecified elements.
  • the term "consisting essentially of” refers to those elements required for a given embodiment. The term permits the presence of additional elements that do not materially affect the basic and novel or functional characteristic(s) of that embodiment of the disclosure.
  • compositions, methods, and respective components thereof as described herein, which are exclusive of any element not recited in that description of the embodiment.
  • antibody herein is used in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity.
  • an "antibody fragment” refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds.
  • antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab')2; diabodies; linear antibodies; single-chain antibody molecules (e.g. scFv); and multispecific antibodies formed from antibody fragments.
  • prophylactic agent refers to CD3 binding molecules such as tephzumab which can be used in the prevention, treatment, management or amelioration of one or more symptoms of T1D.
  • onset of disease with reference to Type-1 diabetes refers to a patient meeting the criteria established for diagnosis of Type-1 diabetes by the American Diabetes Association (see, Mayfield et al., 2006, Am. Fam. Physician 58: 1355-1362).
  • the terms “prevent”, “preventing” and “prevention” refer to the prevention of the onset of one or more symptoms of T1D in a subject resulting from the administration of a prophy lactic or therapeutic agent.
  • a “protocol” includes dosing schedules and dosing regimens.
  • the protocols herein are methods of use and include prophylactic and therapeutic protocols.
  • a “dosing regimen” or “course of treatment” may include administration of several doses of a therapeutic or prophylactic agent over 1 to 20 days.
  • the terms “subject” and “patient” are used interchangeably.
  • the terms “subject” and “subjects” refer to an animal, preferably a mammal including a non-primate (e.g., a cow, pig, horse, cat, dog, rat, and mouse) and a primate (e.g., a monkey or a human), and more preferably a human.
  • a non-primate e.g., a cow, pig, horse, cat, dog, rat, and mouse
  • a primate e.g., a monkey or a human
  • prophylactically effective amount refers to that amount of teplizumab sufficient to result in the delay or prevention of the development, recurrence or onset of one or more symptoms of T1D.
  • a prophylactically effective amount preferably refers to the amount of teplizumab that delays a subject's onset of T1D by at least 20%, by at least 25%, by at least 30%, by at least 35%, by at least 40%, by at least 45%, by at least 50%, by at least 55%, by at least 60%, by at least 65%, by at least 70%, by at least 75%, by at least 80%, by at least 85%, by at least 90%, by at least 95%.
  • anti-CD3 antibody and “an antibody that binds to CD3” refer to an antibody or antibody fragment that is capable of binding cluster of differentiation 3 (CD3) with sufficient affinity such that the antibody is useful as a prophylactic, diagnostic and/or therapeutic agent in targeting CD3.
  • the extent of binding of an anti-CD3 antibody to an unrelated, non-CD3 protein is less than about 10% of the binding of the antibody to CD3 as measured, e.g., by a radioimmunoassay (RIA).
  • RIA radioimmunoassay
  • an antibody that binds to CD3 has a dissociation constant (Kd) of ⁇ 1 pM, ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM (e.g. 10’ 8 M or less, e.g. from 10’ 8 M to 10’ 13 M, e.g., from 10’ 9 M to 10’ 13 M).
  • Kd dissociation constant
  • an anti-CD3 antibody binds to an epitope of CD3 that is conserved among CD3 from different species.
  • the anti-CD3 antibody can be ChAglyCD3 (otelixizumab).
  • Otelixizumab is a humanized Fc nonbinding anti-CD3, which was evaluated initially in phase 2 studies by the Belgian Diabetes Registry (BDR) and then developed by Tolerx, which then partnered with GSK to conduct the phase 3 DEFEND new onset T1D trials (NCT00678886, NCT01123083, NCT00763451).
  • Otelixizumab is administered IV with infusions over 8 days. See, e.g., Wiczling et al., J. Clin. Pharmacol. 50 (5) (May 2010) 494-506; Keymeulen et al., N Engl J Med.
  • the anti-CD3 antibody can be visilizumab (also called HuM291 ; Nuvion).
  • Visilizumab is a humanized anti-CD3 monoclonal antibody characterized by a mutated IgG2 isotype, lack of binding to Fey receptors, and the ability to induce apoptosis selectively in activated T cells. It was evaluated in patients in graft-versus-host disease (NCT00720629; NCT00032279) and in ulcerative colitis (NCT00267306) and Crohn’s Disease (NCT00267709). See, e.g., Sandborn et al., Gut 59 (11) (Nov 2010) 1485-1492, incorporated herein by reference.
  • the anti-CD3 antibody can be foralumab, a fully human anti- CD3 monoclonal antibody being developed by Tiziana Life Sciences, PLC in NASH and T2D (NCT03291249). See, e.g., Ogura et al., Clin Immunol, 2017;183:240-246; Ishikawa et al., Diabetes. 2007;56(8):2103-9; Wuetal., J Immunol, 2010;185(6):3401-7; all incorporated herein by reference.
  • the anti-CD3 antibody can be teplizumab.
  • Teplizumab also known as hOKT3yl(Ala-Ala) (containing an alanine at positions 234 and 235) is an anti-CD3 antibody that had been engineered to alter the function of the T lymphocytes that mediate the destruction of the insulin-producing beta cells of the islets of the pancreas.
  • Teplizumab binds to an epitope of the CD3s chain expressed on mature T cells and by doing so changes their function. Sequences and compositions of teplizumab are disclosed in U.S. Patent Nos. 6,491,916; 8,663,634; and 9,056,906, each incorporated herein by reference in its entirety. The full sequences of light and heavy chains are set forth below. Bolded portions are the complementarity determining regions.
  • Teplizumab Heavy Chain (SEQ ID NO: 2):
  • compositions compnse a prophylactically effective amount of an anti-CD3 antibody, and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant (e.g., Freund's adjuvant (complete and incomplete)), excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like (See, for example. Handbook of Pharmaceutical Excipients, Arthur H. Kibbe (ed., 2000, which is incorporated by reference herein in its entirety), Am. Pharmaceutical Association, Washington, D.C.
  • compositions can also contain minor amounts of weting or emulsifying agents, or pH buffering agents.
  • These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained release formulations and the like.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E. W. Martin.
  • Such compositions will contain a prophylactically or therapeutically effective amount of a prophylactic or therapeutic agent preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient.
  • the formulation should suit the mode of administration.
  • the pharmaceutical compositions are sterile and in suitable form for administration to a subject, preferably an animal subject, more preferably a mammalian subject, and most preferably a human subject.
  • the pharmaceutical compositions may be desirable to administer the pharmaceutical compositions locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion, by injection, or by means of an implant, the implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • the implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • care must be taken to use materials to which the anti-CD3 antibody does not absorb.
  • the composition can be delivered in a vesicle, in particular a liposome (see Langer, Science 249: 1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353- 365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.).
  • a liposome see Langer, Science 249: 1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353- 365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.).
  • the composition can be delivered in a controlled release or sustained release system.
  • a pump may be used to achieve controlled or sustained release (see Langer, supra; Sefton, 1987, CRC Crit. Ref. Biomed. Eng. 14:20; Buchwald et al., 1980, Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med. 321 :574).
  • polymeric materials can be used to achieve controlled or sustained release of the antibodies of the invention or fragments thereof (see e.g., Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla.
  • polymers used in sustained release formulations include, but are not limited to, poly(2-hydroxy ethyl methacrylate), poly(methyl methacrylate), poly(acrylic acid), poly(ethylene-co-vinyl acetate), poly(methacrylic acid), polyglycolides (PLG), polyanhydrides, poly(N-vinyl pyrrolidone), poly(vinyl alcohol), polyacrylamide, poly(ethylene glycol), polylactides (PLA), poly(lactide-co-glycolides) (PLGA), and polyorthoesters.
  • the polymer used in a sustained release formulation is inert, free of leachable impurities, stable on storage, sterile, and biodegradable.
  • a controlled or sustained release system can be placed in proximity of the therapeutic target, i.e., the lungs, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
  • Controlled release systems are discussed in the review by Langer (1990, Science 249: 1527-1533). Any technique known to one of skill in the art can be used to produce sustained release formulations comprising one or more antibodies of the invention or fragments thereof. See, e.g., U.S. Pat. No. 4,526,938; PCT Publication No. WO 91/05548; PCT Publication No. WO 96/20698; Ning et al., 1996, Radiotherapy & Oncology 39:179-189; Song et al., 1995, PDA Journal of Pharmaceutical Science & Technology 50:372-397; Cleek et al., 1997, Pro. Infl. Symp. Control. Rel. Bioact. Mater. 24:853-854; and Lam et al., 1997, Proc. Infl. Symp. Control Rel. Bioact. Mater. 24:759-760, each of which is incorporated herein by reference in its entirety.
  • a pharmaceutical composition can be formulated to be compatible with its intended route of administration.
  • routes of administration include, but are not limited to, parenteral, e.g., intravenous, intradermal, subcutaneous, oral, intranasal (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration.
  • the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous, subcutaneous, intramuscular, oral, intranasal or topical administration to human beings.
  • a pharmaceutical composition is formulated in accordance with routine procedures for subcutaneous administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lignocamne to ease pain at the site of the injection.
  • compositions may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • the disclosure provides dosage forms that permit administration of the anti-CD3 antibody continuously over a period of hours or days (e.g., associated with a pump or other device for such delivery), for example, over a period of 1 hour, 2 hours, 3 hours, 4 hours, 6 hours, 8 hours, 10 hours, 12 hours, 16 hours, 20 hours, 24 hours, 30 hours, 36 hours, 4 days, 5 days, 7 days, 10 days or 14 days.
  • the invention provides dosage forms that permit administration of a continuously increasing dose, for example, increasing from 51 ug/m 2 /day to 826 ug/m 2 /day over a period of 24 hours, 30 hours, 36 hours, 4 days, 5 days, 7 days, 10 days or 14 days.
  • compositions can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, tri ethyl amine, 2-ethylamino ethanol, histidine, procaine, etc.
  • the ingredients of the compositions disclosed herein are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle or bag containing sterile pharmaceutical grade water or saline.
  • the composition is in solution.
  • the composition is packaged into a vial (e.g. sterile glass vial of 0.9% sodium chloride injection) or into an infusion bag (e.g. Polyvinylchloride (PVC) infusion bag of 0.9% sodium chloride injection).
  • the vial is for single-dose.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • the disclosure provides that the anti-CD3 antibodies, or pharmaceutical compositions thereof, can be packaged in a hermetically sealed container such as an ampoule or sachette indicating the quantity of the agent.
  • the anti-CD3 antibody, or pharmaceutical compositions thereof is supplied as a dry sterilized lyophilized powder or water free concentrate in a hermetically sealed container and can be reconstituted, e.g., with water or saline to the appropriate concentration for administration to a subject.
  • the anti-CD3 antibody, or pharmaceutical compositions thereof is supplied as a dry sterile lyophilized powder in a hermetically sealed container at a unit dosage of at least 5 mg, more preferably at least 10 mg, at least 15 mg, at least 25 mg, at least 35 mg, at least 45 mg, at least 50 mg, at least 75 mg, or at least 100 mg.
  • the lyophilized prophylactic agents, or pharmaceutical compositions herein should be stored at between 2 °C and 8 °C in its original container and the prophylactic or therapeutic agents, or pharmaceutical compositions of the invention should be administered within 1 week, preferably within 5 days, within 72 hours, within 48 hours, within 24 hours, within 12 hours, within 6 hours, within 5 hours, within 3 hours, or within 1 hour after being reconstituted.
  • the pharmaceutical composition is supplied in liquid form in a hermetically sealed container indicating the quantity and concentration of the agent.
  • the liquid fonn of the administered composition is supplied in a hermetically sealed container at least 0.25 mg/ml, more preferably at least 0.5 mg/ml, at least 1 mg/ml, at least 2.5 mg/ml, at least 5 mg/ml, at least 8 mg/ml, at least 10 mg/ml, at least 15 mg/ml, at least 25 mg/ml, at least 50 mg/ml, at least 75 mg/ml or at least 100 mg/ml.
  • the liquid form should be stored at between 2 °C and 8 °C in its original container.
  • the disclosure provides that the composition of the invention is packaged in a hennetically sealed container such as an ampoule or sachette indicating the quantity of the anti-CD3 antibody.
  • compositions may, if desired, be presented in a pack or dispenser device that may contain one or more unit dosage forms containing the active ingredient.
  • the pack may, for example, comprise metal or plastic foil, such as a blister pack.
  • the amount of the composition of the invention which will be effective in the prevention or amelioration of one or more symptoms associated with T1D can be determined by standard clinical techniques.
  • the precise dose to be employed in the formulation will also depend on the route of administration and the seriousness of the condition, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the present disclosure encompasses administration of antihuman CD3 antibodies such as teplizumab to individuals predisposed to develop type 1 diabetes or with pre-clinical stages of type 1 diabetes, but who do not meet the diagnosis criteria as established by the American Diabetes Association or the Immunology of Diabetes Society to prevent or delay the onset of type 1 diabetes and/or to prevent or delay the need for administration of insuhn to such patients.
  • antihuman CD3 antibodies such as teplizumab
  • high-risk factors for identification of predisposed subjects include having first or second degree relatives with diagnosed type-1 diabetes, an impaired fasting glucose level (e.g., at least one determination of a glucose level of 100-125 mg/dl after fasting (8 hours with no food)), an impaired glucose tolerance in response to a 75 g OGTT (e.g., at least one determination of a 2-hr glucose level of 140-199 mg/dl in response to a 75 g OGTT), an HLA type of DR3, DR4 or DR7 in a Caucasian, an HLA type of DR3 or DR4 in a person of African descent, an HLA type of DR3, DR4 or DR9 in a person of Japanese descent, exposure to viruses (e.g., coxsackie B virus, enteroviruses, adenoviruses, rubella, cytomegalovirus, Epstein-Barr vims), a positive diagnosis according to art accepted criteria of at least one other autoimmune disorder (e.
  • viruses
  • the subject identified as predisposed to developing type 1 diabetes has at least one of the risk factors described herein and/or as known in the art.
  • the present disclosure also encompasses identification of subjects predisposed to development of type 1 diabetes, wherein the subject presents a combination of two or more, three or more, four or more, or more than five of the risk factors disclosed herein or known in the art.
  • the non-diabetic subject is an adult or a pediatric subject.
  • the pediatric subject is 8 years old or older.
  • the pediatric subject is 7 years old or older.
  • the pediatric subject is 6 years old or older.
  • the pediatric subject is 5 years old or older.
  • the pediatric subject is 4 years old or older.
  • the pediatric subject is 3 years old or older. In some embodiments, the pediatric subject is 2 years old or older. In some embodiments, the pediatric subject is 1 year old or older. In some embodiments, the pediatric subject is an infant. In some embodiments, the infant is 12 months old or less, 11 months old or less, 10 months old of less, 9 months old or less, 8 months old or less, 7 months old or less, 6 months old or less, 5 months old or less, 4 months old or less, 3 months old or less, 2 months old or less, I month old or less.
  • Serum autoantibodies associated with type 1 diabetes or with a predisposition for the development of type 1 diabetes are islet-cell autoantibodies (e.g., anti-ICA512 autoantibodies), glutamic acid decarbamylase autoantibodies (e.g., anti-GAD65 autoantibodies), IA2 antibodies, ZnT8 antibodies and/or anti-insulin autoantibodies.
  • islet-cell autoantibodies e.g., anti-ICA512 autoantibodies
  • glutamic acid decarbamylase autoantibodies e.g., anti-GAD65 autoantibodies
  • IA2 antibodies e.g., ZnT8 antibodies and/or anti-insulin autoantibodies.
  • the invention encompasses the treatment of an individual with detectable autoantibodies associated with a predisposition to the development of type 1 diabetes or associated with early stage type 1 diabetes (e.g., anti-IA2, anti-ICA512, anti-GAD or antiinsulin autoantibodies), wherein the individual has not been diagnosed with type 1 diabetes and/or is a first or second degree relative of a type-1 diabetic.
  • the presence of the autoantibodies is detected by ELISA, electrochemoluminescence (ECL), radioassay (see, e.g., Yu et al., 1996, J. Clin. Endocrinol. Metab.
  • P-cell function prior to, during, and after therapy may be assessed by methods described herein or by any method known to one of ordinary skill in the art.
  • DCCT Diabetes Control and Complications Trial
  • HA1 and HAlc percentage glycosylated hemoglobin
  • characterization of daily insulin needs, C-peptide levels/response, hypoglycemic episodes, and/or FPIR may be used as markers of p-cel 1 function or to establish a therapeutic index (See Keymeulen et al, 2005, N. Engl. J. Med.
  • FPIR is calculated as the sum of insulin values at 1 and 3 minutes post IGTT, which are performed according to Islet Cell Antibody Register User's Study protocols (see, e.g., Bingley et al, 1996, Diabetes 45: 1720-1728 and McCulloch et al., 1993, Diabetes Care 16:911-915).
  • the individuals predisposed to develop T1D can be a nondiabetic subject who is a relative of a patient with T1D.
  • the non-diabetic subject has 2 or more diabetes-related autoantibodies selected from islet cell antibodies (ICA), insulin autoantibodies (IAA), and antibodies to glutamic acid decarboxylase (GAD), tyrosine phosphatase (IA-2/ICA512) or ZnT8.
  • ICA islet cell antibodies
  • IAA insulin autoantibodies
  • GAD glutamic acid decarboxylase
  • IA-2/ICA512 tyrosine phosphatase
  • the non-diabetic subj ect has abnormal glucose tolerance on oral glucose tolerance test (OGTT).
  • OGTT oral glucose tolerance test
  • Abnormal glucose tolerance on OGTT is defined as a fasting glucose level of 110-125 mg/dL, or 2 hour plasma of > 140 and ⁇ 200 mg/dL, or an intervening glucose value at 30, 60, or 90 minutes on OGTT > 200 mg/dL.
  • CGM glucose monitoring system
  • the non-diabetic subject who will respond to the anti-CD3 antibody such as teplizumab does not have antibodies against ZnT8.
  • such non-diabetic subject is HLA-DR4+ and is not HLA-DR3+.
  • such non- diabetic subject who will respond to the anti-CD3 antibody such as teplizumab demonstrates an increase, following administration (e.g., after 1 month, after 2 months, after 3 months, or longer or shorter), in the frequency (or relative amount) of TIGIT+KLRG1+CD8+ T-cells (e.g., by flow cytometry) in peripheral blood mononuclear cells.
  • the methods provided herein comprise administering an antihuman CD3 antibodies such as teplizumab to a subject in need thereof with Stage 2 type 1 diabetes.
  • the subject is an adult.
  • the subject is a pediatric subject.
  • the pediatric subject is 8 years old or older (e.g. 8 years old, 9 years old, 10 years old, 11 years old, 12 years old, 13 years old, 14 years old, 15 years old, 16 years old, 17 years old, 18 years old or older).
  • administration of the anti-human CD3 antibodies such as teplizumab delay the onset of Stage 3 Type 1 diabetes.
  • the subject has Stage 2 type 1 diabetes.
  • the subject that has dysglycemia without overt hyperglycemia has at least two positive pancreatic islet cell autoantibodies. See ADA and ISPAD guidelines which define classification and diagnosis of Stage 2 T1D in American Diabetes Association Professional Practice Committee- Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes-2022. Diabetes Care. 2022 Jan l;45(Suppl 1):S 17-S38); and in Besser REJ, Bell KJ, Couper JJ, Ziegler AG, Wherrett DK, Knip M, Speake C, Casteels K, Driscoll KA, Jacobsen L, Craig ME, Haller MJ.
  • the subject has abnormal glucose tolerance on oral glucose tolerance test (OGTT).
  • OGTT oral glucose tolerance test
  • the subject in need thereof meets criteria for a diagnosis of Stage 2 type 1 diabetes does not have type 2 diabetes.
  • the methods provided herein comprise confirming that the subject who has dysglycemia without overt hyperglycemia has Stage 2 type 1 diabetes by documenting at least two positive pancreatic islet cell autoantibodies.
  • the methods provided herein further comprise confirming that the subject does not have type 2 diabetes.
  • subjects having Stage 2 type 1 diabetes have both of the following:
  • pancreatic islet autoantibodies Two or more of the following pancreatic islet autoantibodies:
  • Glutamic acid decarboxylase 65 (GAD) autoantibodies
  • IAA Insulin autoantibody
  • Insulinoma-associated antigen 2 autoantibody IA-2A
  • Zinc transporter 8 autoantibody ZnT8A
  • Dysglycemia e g. on oral glucose tolerance testing.
  • Stage 2 type 1 diabetes can be diagnosed in other ways known to those skilled in the art.
  • aspects of the disclosure relate to methods of preventing or delaying onset of Stage 3 type 1 diabetes (T1D).
  • the subject is an adult.
  • the subject is a child 1 year old, 2 years old, 3 years old or older, 4 years old and older, 5 years old and older, 6 years old and older, 7 years old and older, 8 years old and older, 9 years old and older, 10 years old and older, 11 years old and older, 12 years old and older, 13 years old and older, 14 years old and older, 15 years old and older, 16 years old and older, 17 years old and older, 18 years old and older.
  • the subject is a infant ( 12 months old or less, 11 months old or less, 10 months old or less, 9 months old or less, 8 months old or less, 7 months old or less, 6 months old or less, 5 months old or less, 4 months old or less, 3 months old or less, 2 months old or less, 1 month old or less (e.g. initial weeks of life)).
  • the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at a cumulative dose greater than 10,000 micrograms/meter squared (pg/m 2 ).
  • the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 9,500 to about 14,000 pg/m 2 , from about 9,500 to about 13,500 pg/m 2 , from about 9,500 to about 13,000 pg/ m 2 , from about 9,500 to about 12,500 pg/m 2 , from about 9,500 to about 12,000 pg/m 2 , from about 9,500 to about 11,500 pg/ m 2 , from about 9,500 to about 11,000 pg/m 2 , from about 9,500 to about 10,500 pg/m 2 , from about 9,500 to about 10,000 pg/m 2 .
  • SC subcutaneous
  • IV intravenous
  • the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 10,000 to about 14,000 pg/m 2 , from about 10,000 to about 13,500 pg/ m 2 , from about 10,000 to about 13,000 pg/m 2 , from about 10,000 to about 12,500 pg/m 2 , from about 10,000 to about 12,000 pg/m 2 , from about 10,000 to about 11,500 pg/m 2 , from about 10,000 to about 11,000 pg/ m 2 , from about 10,000 to about 10,500 pg/m 2 .
  • SC subcutaneous
  • IV intravenous
  • the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 10,500 to about 14,000 pg/m 2 , from about 10,500 to about 13,500 pg/m 2 , from about 10,500 to about 13,000 pg/m 2 , from about 10,500 to about 12,500 pg/m 2 , from about 10,500 to about 12,000 pg/m 2 , from about 10,500 to about 11,500 pg/ m 2 , from about 10,500 to about 11,000 pg/m 2 .
  • SC subcutaneous
  • IV intravenous
  • the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 11,000 to about 14,000 pg/m 2 , from about 11,000 to about 13,500 pg/m 2 , from about 11,000 to about 13,000 pg/m 2 , from about 11,000 to about 12,500 pg/m 2 , from about 11,000 to about 12,000 pg/m 2 , from about 11,000 to about 11,500 pg/m 2 .
  • SC subcutaneous
  • IV intravenous
  • the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 11,000 to about 14,000 pg/m 2 , from about 11,000 to about 13,500 pg/ m 2 , from about 11,000 to about 13,000 pg/m 2 , from about 11,000 to about 12,500 pg/m 2 , from about 11,000 to about 12,000 pg/m 2 , from about 11,000 to about 11,500 pg/m 2 .
  • SC subcutaneous
  • IV intravenous
  • the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 11,500 to about 14,000 pg/m 2 , from about 11,500 to about 13,500 pg/m 2 , from about 11,500 to about 13,000 pg/m 2 , from about 11,500 to about 12,500 pg/m 2 , from about 11,500 to about 12,000 pg/m 2 .
  • SC subcutaneous
  • IV intravenous
  • the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 12,000 to about 14,000 pg/m 2 , from about 12,000 to about 13,500 pg/m 2 , from about 12,000 to about 13,000 pg/m 2 , from about 12,000 to about 12,500 pg/m 2 .
  • the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 12,500 to about 14,000 pg/m 2 , from about 12,500 to about 13,500 pg/m 2 , from about 12,500 to about 13,000 pg/m 2 .
  • the prophy tactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 13,000 to about 14,000 pg/m 2 , from about 13,000 to about 13,500 pg/m 2 .
  • the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 13,500 pg/m 2 to about 14,000 pg/m 2 .
  • the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at about 10,000 pg/m 2 , 10,500 pg/m 2 , 11,000 pg/m 2 , 11.500 pg/m 2 , 12,000 pg/m 2 , 12,500 pg/m 2 , 13,000 pg/m 2 , 13,500 pg/m 2 , or 14,000 pg/m 2 .
  • the anti-CD3 antibody is or comprises teplizumab.
  • the prophylactically effective amount of the antibody comprises a 10 to 14 days course of intravenous (IV) infusion of teplizumab at from about 10,000 to about 14,000 pg/m 2 , from about 10,000 to about 13,500 pg/m 2 , from about 10,000 to about 13,000 pg/m 2 , from about 10,000 to about 12,500 pg/m 2 , from about 10,000 to about 12,000 pg/m 2 , from about 10,000 to about 11,500 pg/ m 2 , from about 10,000 to about 11,000 pg/m 2 , from about 10,000 to about 10,500 pg/m 2 .
  • IV intravenous
  • the prophylactically effective amount of the antibody comprises a 10 to 14 days course of intravenous (IV) infusion of teplizumab at from about 10,500 to about 14,000 pg/m 2 , from about 10,500 to about 13,500 pg/m 2 , from about 10,500 to about 13,000 pg/m 2 , from about 10,500 to about 12,500 pg/m 2 , from about 10,500 to about 12,000 pg/m 2 , from about 10,500 to about 11,500 pg/m 2 , from about 10,500 to about 11,000 pg/m 2 .
  • IV intravenous
  • the prophylactically effective amount of the antibody comprises a 10 to 14 days course of intravenous (IV) infusion of teplizumab at from about 11 ,000 to about 14,000 pg/m 2 , from about 11,000 to about 13,500 pg/m 2 , from about 11,000 to about 13,000 pg/m 2 , from about 11,000 to about 12,500 pg/m 2 , from about 11,000 to about 12,000 pg/m 2 , from about 11,000 to about 11,500 pg/m 2 .
  • IV intravenous
  • the prophylactically effective amount of the antibody comprises 10 to 14 days course of intravenous (IV) infusion of teplizumab at from about 11,000 to about 14,000 pg/m 2 , from about 11,000 to about 13,500 pg/m 2 , from about 11,000 to about 13,000 pg/m 2 , from about 11,000 to about 12,500 pg/m 2 , from about 11,000 to about 12,000 pg/m 2 , from about 11,000 to about 11,500 pg/m 2 .
  • IV intravenous
  • the prophylactically effective amount of the antibody comprises 10 to 14 days course of intravenous (IV) infusion of teplizumab at from about 11,500 to about 14,000 pg/m 2 , from about 11,500 to about 13,500 gg/m 2 , from about 11,500 to about 13,000 gg/m 2 , from about
  • the prophylactically effective amount of the antibody comprises 10 to 14 days course of intravenous (IV) infusion of tephzumab at from about 12,000 to about 14,000 gg/m 2 , from about 12,000 to about 13,500 gg/m 2 , from about 12,000 to about 13,000 gg/m 2 , from about 12,000 to about 12,500 gg/m 2 .
  • the prophylactically effective amount of the antibody comprises 10 to 14 days course of intravenous (IV) infusion of teplizumab at from about 12,500 to about 14,000 gg/m 2 , from about 12,500 to about 13,500 gg/m 2 , from about
  • the prophylactically effective amount of the antibody comprises 10 to 14 days course of intravenous (IV) infusion of teplizumab at from about 13,000 to about 14,000 gg/m 2 , from about 13,000 to about 13,500 gg/m 2 . In some embodiments, the prophylactically effective amount of the antibody comprises 10 to 14 days course of intravenous (IV) infusion of teplizumab at from about 13,500 gg/m 2 to about 14,000 gg/m 2 .
  • the prophylactically effective amount of the antibody comprises 10 to 14 days course of intravenous (IV) infusion of teplizumab at about 10,000 gg/ m 2 , 10,500 gg/m 2 , 11,000 gg/m 2 , 11.500 gg/m 2 , 12,000 gg/m 2 , 12,500 gg/m 2 , 13,000 gg/m 2 , 13,500 gg/m 2 , or 14,000 gg/m 2 .
  • the anti-CD3 antibody is or comprises teplizumab.
  • the method comprises administering to the non-diabetic subj ect who is at risk for T1D a 14-day course IV infusion at about 60 gg/m 2 , about 125 gg/m 2 , about 250 gg/m 2 , and about 500 gg/m 2 , on days 1-4, respectively, and a dose of about 1,000 gg/m 2 on each of days 5-14. In some embodiments, the cumulative dose is about 10,935 gg/m 2 .
  • the method comprises administering to the non-diabetic subj ect who is at risk for T1D a 14-day course IV infusion at about 60 gg/m 2 , about 125 gg/m 2 , about 250 gg/m 2 , and about 500 gg/m 2 , on days 1-4, respectively, and a dose of about 1,030 gg/m 2 on each of days 5-14.
  • the cumulative dose is about 11,235 gg/m 2 .
  • the method comprises administering to the non-diabetic subject who is at risk for T1D a 14-day course IV infusion at about 100 gg/m 2 , about 425 gg/m 2 , about 850 gg/m 2 , and about 850 gg/m 2 , on days 1-4, respectively, and a dose of about 1,000 gg/m 2 on each of days 5-14.
  • the cumulative dose is about 12,225 gg/m 2 .
  • the method comprises administering to the non-diabetic subject who is at risk for T1D a 14-day course IV infusion at about 65 gg/m 2 , about 125 gg/m 2 , about 250 pg/m 2 , and about 500 pg/m 2 , on days 1-4, respectively, and a dose of about 1,070 pg/m 2 on each of days 5-14.
  • the cumulative dose is about 11,640 pg/m 2 .
  • the method comprises administering to a subject in need thereof a 14-day course IV infusion, wherein the daily dose is at about 65 pg/m 2 , about 125 pg/m 2 , about 250 pg/m 2 , and about 500 pg/m 2 , on days 1-4, respectively, and at about 1,030 pg/m 2 on each of days 5-14.
  • the cumulative dose is about 11,240 pg/m 2 .
  • the method comprises administering to a subject in need thereof a 14-day course IV infusion, wherein the daily dose is at about 60 pg/m 2 , about 125 pg/m 2 , about 250 pg/m 2 , and about 500 pg/m 2 , on days 1-4, respectively, and at about 1,000 pg/m 2 on each of days 5-14.
  • the cumulative dose is about 10,935 pg/m 2 .
  • the method comprises administering to a subject in need thereof a 14-day course IV infusion, wherein the daily dose is at about 60 pg/m 2 , about 125 pg/m 2 , about 250 pg/m 2 , and about 500 pg/m 2 , on days 1-4, respectively, and at about 1,030 pg/m 2 on each of days 5-14.
  • the cumulative dose is about 11,235 pg/m 2 .
  • the method comprises administering to subject in need therof a 14-day course IV infusion, wherein the daily dose is at about 100 pg/m 2 , about 425 pg/m 2 , about 850 pg/m 2 , and about 850 pg/m 2 , on days 1-4, respectively, and at about 1,000 pg/m 2 on each of days 5-14.
  • the cumulative dose is about 12,225 pg/m 2 .
  • the method comprises administering to a subject in need thereof a 14-day course IV infusion, wherein the daily dose is at about 65 pg/m 2 , about 125 pg/m 2 , about 250 pg/m 2 , and about 500 pg/m 2 , on days 1-4, respectively, and a dose of about 1,070 pg/m 2 on each of days 5-14.
  • the cumulative dose is about 11,640 pg/m 2 .
  • the method comprises administering to a subject in need thereof a 14-day course IV infusion, wherein the daily dose is at about 65 pg/m 2 , about 125 pg/m 2 , about 250 pg/m 2 , and about 500 pg/m 2 , on days 1-4, respectively, and at about 1,030 pg/m 2 on each of days 5-14.
  • the cumulative dose is about 11,240 pg/m 2 .
  • the anti-CD3 antibody daily administration is missed, and dosing resumes by administering all remaining doses on consecutive days to complete the 14-day treatment course.
  • the prophylactically effective amount delays median time to clinical diagnosis of T1D by at least 50%, at least 80%, or at least 90%, or at least 12 months, at least 18 months, at least 24 months, at least 36 months, at least 48 months, or at least 60 months, or longer.
  • the course of dosing with the anti-CD3 antibody such as teplizumab can be repeated at 2 month, 4 month, 6 month, 8 month, 9 month, 10 month, 12 month, 15 month, 18 month, 24 month, 30 month, or 36 month intervals.
  • efficacy of the treatment with the anti-CD3 antibody such as teplizumab is determined as described herein, or as is known in the art, at 2 months, 4 months, 6 months, 9 months, 12 months, 15 months, 18 months, 24 months, 30 months, or 36 months subsequent to the previous treatment.
  • a subject is administered one or more unit doses of approximately 0.5-50 pg/kg, approximately 0.5-40 pg/kg, approximately 0.5-30 pg/kg, approximately 0.5-20 pg/kg, approximately 0.5-15 pg/kg, approximately 0.5-10 pg/kg, approximately 0.5-5 pg/kg, approximately 1-5 pg/kg, approximately 1-10 pg/kg, approximately 20-40 pg/kg, approximately 20-30 pg/kg, approximately 22-28 pg/kg or approximately 25-26 pg/kg of the anti-CD3 antibody such as teplizumab to prevent, treat, ameliorate, mitigate, relieve, lighten, postpone, stop or halt one or more symptoms of T1D.
  • the anti-CD3 antibody such as teplizumab
  • a subject is administered one or more unit doses of about 200 pg/kg, 178 pg/kg, 180 pg/kg, 128 pg/kg, 100 pg/kg, 95 pg/kg, 90 pg/kg, 85 pg/kg, 80 pg/kg, 75 pg/kg, 70 pg/kg, 65 pg/kg, 60 pg/kg, 55 pg/kg, 50 pg/kg, 45 pg/kg, 40 pg/kg, 35 pg/kg, 30 pg/kg, 26 pg/kg, 25 pg/kg, 20 pg/kg, 15 pg/kg, 13 pg/kg, 10 pg/kg, 6.5 pg/kg, 5 pg/kg, 3.2 pg/kg, 3 pg/kg, 2.5 pg/kg, 2 pg/kg, 1.6 pg/kg, 1.5 pg/kg, 1
  • a subject is administered one or more doses of the anti-CD3 antibody such as teplizumab at about 5-1200 pg/m 2 , for example, at about 60-1070 pg/m 2 .
  • a subject is administered one or more unit doses of 1200 pg/m 2 , 1150 pg/m 2 , 1100 pg/m 2 , 1050 pg/m 2 , 1000 pg/m 2 , 950 pg/m 2 , 900 pg/m 2 , 850 pg/m 2 , 800 pg/m 2 , 750 pg/m 2 , 700 pg/m 2 , 650 pg/m 2 , 600 ug/m 2 , 550 pg/m 2 , 500 pg/m 2 , 450 pg/m 2 , 400 pg/m 2 , 350 pg/m 2 , 300 pg/m 2 , 250 pg/m 2 , 200 pg/m 2 , 150 pg/m 2 , 100 pg/m 2 , 50 pg/m 2 , 40 pg/m 2 , 30
  • the subject is administered a treatment regimen comprising one or more doses of a prophylactically effective amount of the anti-CD3 antibody such as teplizumab, wherein the course of treatment is administered over 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days or 14 days.
  • the subject is administered a treatment regimen comprising one or more doses of a prophylactically effective amount of the anti-CD3 antibody such as teplizumab, wherein the course of treatment is administered over 14 days.
  • the treatment regimen comprises administering doses of the prophylactically effective amount every day, every 2nd day, every 3rd day or every 4th day. In some embodiments, the treatment regimen comprises administering doses of the prophylactically effective amount on Monday, Tuesday, Wednesday, Thursday of a given week and not administering doses of the prophylactically effective amount on Friday, Saturday, and Sunday of the same week until 14 doses, 13 doses, 12 doses, 11 doses, 10 doses, 9 doses, or 8 doses have been administered. In some embodiments, the dose administered is the same each day of the regimen.
  • a subject is administered a treatment regimen comprising one or more doses of a prophylactically effective amount of the anti-CD3 antibody such as teplizumab, wherein the prophylactically effective amount is about 200 pg/kg /day, 175 pg/kg/day, 150 pg/kg/day, 125 pg/kg/day, 100 pg/kg/day, 95 pg/kg/day, 90 pg/kg/day, 85 pg/kg/day, 80 pg/kg/day, 75 pg/kg/day, 70 pg/kg/day, 65 pg/kg/day, 60 pg/kg/day, 55 pg/kg/day, 50 pg/kg/day, 45 pg/kg/day, 40 pg/kg/day, 35 pg/kg/day, 30 pg/kg/day, 26 pg/kg/day, 25 pg/kg/day, 20
  • the intravenous dose of about 1200 pg/m 2 or less, 1150 pg/m 2 or less, 1100 pg/m 2 or less, 1050 pg/m 2 or less, 1000 pg/m 2 or less, 950 pg/m 2 or less, 900 pg/m 2 or less, 850 pg/m 2 or less, 800 pg/m 2 or less, 750 pg/m 2 or less, 700 pg/m 2 or less, 650 pg/m 2 or less, 600 gg/m 2 or less, 550 gg/m 2 or less, 500 gg/m 2 or less, 450 gg/m 2 or less, 400 gg/m 2 or less, 350 gg/m 2 or less, 300 gg/m 2 or less, 250 gg/m 2 or less, 200 gg/m 2 or less, 150 gg/m 2 or less, 100 gg/m 2 or less, 50 gg/m 2 or less,
  • the total dosage over the duration of the regimen can be greater than about 9,500, 10,000, 10,500, 11,000, 11,500, 12,000, 12,500, 13,000 or 13,500 ug/m 2 .
  • the total dosage over the duration of the regimen in some embodiments, is from about 9,500 to about 14,000 gg/m 2 , from about 9,500 to about
  • the total dosage over the duration of the regimen in some embodiments, is from about 10,500 to about 14,000 gg/m 2 , from about 10,500 to about 13,500 gg/m 2 , from about
  • the total dosage over the duration of the regimen is about 10,935 gg/m 2 , about 11,235 gg/m 2 , about 11,240 gg/m 2 , about 11,640 gg/m 2 , or about 12,225 gg/m 2
  • the dose escalates over the first fourth, first half or first 2/3 of the doses (e.g., over the first 2, 3, 4, 5, or 6 days of a 10, 12, 14, 16, 18 or 20-day regimen of one dose per day) of the treatment regimen until the daily prophylactically effective amount of the anti-CD3 antibody such as teplizumab is achieved.
  • the dose can escalate over 3 or 4 days of a 14-day regimen.
  • a subject is administered a treatment regimen comprising one or more doses of a prophylactically effective amount of the anti-CD3 antibody such as teplizumab, wherein the prophylactically effective amount is increased by, e g., about 0.01 gg/kg, 0.02 gg/kg, 0.04 gg /kg, 0.05 gg/kg, 0.06 gg/kg, 0.08 gg/kg, 0.1 gg/kg, 0.2 gg/kg, 0.25 gg /kg, 0.5 gg /kg, 0.75 gg /kg, 1 gg /kg, 1.5 gg /kg, 2 gg /kg, 4 gg/kg, 5 gg/kg, 10 gg/kg, 15 gg /kg, 20 gg /kg, 25 gg /kg, 30 gg /kg, 35 gg /kg, 40 gg /kg, 45 gg /kg, 50 gg /kg, 55 gg /kg, 60 gg /
  • a subject is administered a treatment regimen comprising one or more doses of a prophylactically effective amount of the anti-CD3 antibody such as teplizumab, wherein the prophylactically effective amount is increased by a factor of 1.25, a factor of 1.5, a factor of 2, a factor of 2.25, a factor of 2.5, or a factor of 5 until the daily prophylactically effective amount of the anti-CD3 antibody such as teplizumab is achieved.
  • a prophylactically effective amount of the anti-CD3 antibody such as teplizumab
  • a subject is intramuscularly administered one or more doses of a about 200 gg/kg or less, preferably about 175 gg/kg or less, 150 gg/kg or less, 125 gg/kg or less, 100 gg/kg or less, 95 gg/kg or less, 90 gg/kg or less, 85 gg/kg or less, 80 gg/kg or less, 75 gg/kg or less, 70 gg/kg or less, 65 gg/kg or less, 60 gg/kg or less, 55 gg/kg or less, 50 gg/kg or less, 45 gg/kg or less, 40 gg/kg or less, 35 gg/kg or less, 30 gg/kg or less, 25 gg/kg or less, 20 gg/kg or less, 15 gg/kg or less, 10 gg/kg or less, 5 gg/kg or less, 2.5 gg/kg or less, 2 gg/kg or less, 1.5 gg/kg or less, 1 gg/kg or less
  • a subject is subcutaneously administered one or more doses of a about 200 ug/kg or less, preferably about 175 gg/kg or less, 150 gg/kg or less, 125 gg/kg or less, 100 gg/kg or less, 95 gg/kg or less, 90 gg/kg or less, 85 gg/kg or less, 80 gg/kg or less, 75 gg/kg or less, 70 gg/kg or less, 65 gg/kg or less, 60 gg/kg or less, 55 gg/kg or less, 50 gg/kg or less, 45 gg/kg or less, 40 gg/kg or less, 35 gg/kg or less, 30 gg/kg or less, 25 gg/kg or less, 20 gg/kg or less, 15 gg/kg or less, 10 gg/kg or less, 5 gg/kg or less, 2.5 gg/kg or less, 2 gg/kg or less, 1.5 gg/kg or less, 1 gg/kg or less
  • a subject is intravenously administered one or more doses of a about 100 ug/kg or less, preferably about 95 ug/kg or less, 90 ug/kg or less, 85 ug/kg or less, 80 ug/kg or less, 75 ug/kg or less, 70 ug/kg or less, 65 ug/kg or less, 60 ug/kg or less, 55 ug/kg or less, 50 ug/kg or less, 45 ug/kg or less, 40 ug/kg or less, 35 ug/kg or less, 30 ug/kg or less, 25 ug/kg or less, 20 ug/kg or less, 15 ug/kg or less, 10 ug/kg or less, 5 ug/kg or less, 2.5 ug/kg or less, 2 ug/kg or less, 1.5 ug/kg or less, 1 ug/kg or less, 0.5 ug/kg or less,
  • the intravenous dose of about 100 pg/kg or less, 95 pg/kg or less, 90 pg/kg or less, 85 pg/kg or less, 80 pg/kg or less, 75 pg/kg or less, 70 pg/kg or less, 65 u pg/kg or less, 60 pg/kg or less, 55 pg/kg or less, 50 pg/kg or less, 45 pg/kg or less, 40 pg/kg or less, 35 pg/kg or less, 30 pg/kg or less, 25 pg/kg or less, 20 pg/kg or less, 15 pg/kg or less, 10 pg/kg or less, 5 pg/kg or less, 2.5 pg/kg or less, 2 pg/kg or less, 1.5 pg/kg or less, 1 pg/kg or less, 0.5 pg/kg or less, or 0.2 pg/kg or less of the anti-CD
  • a subject is orally administered one or more doses of a about 100 pg/kg or less, preferably about 95 pg/kg or less, 90 pg/kg or less, 85 pg/kg or less, 80 pg/kg or less, 75 pg/kg or less, 70 pg/kg or less, 65 pg/kg or less, 60 pg/kg or less, 55 pg/kg or less, 50 pg/kg or less, 45 pg/kg or less, 40 pg/kg or less, 35 pg/kg or less, 30 pg/kg or less, 25 pg/kg or less, 20 pg/kg or less, 15 pg/kg or less, 10 pg/kg or less, 5 pg/kg or less, 2.5 pg/kg or less, 2 pg/kg or less, 1.5 pg/kg or less, 1 pg/kg or less, 0.5 pg/kg or less, or
  • the oral dose of about 100 pg/kg or less, 95 pg/kg or less, 90 pg/kg or less, 85 pg/kg or less, 80 pg/kg or less, 75 pg/kg or less, 70 pg/kg or less, 65 pg/kg or less, 60 pg/kg or less, 55 pg/kg or less, 50 pg/kg or less, 45 pg/kg or less, 40 pg/kg or less, 35 pg/kg or less, 30 pg/kg or less, 25 pg/kg or less, 20 pg/kg or less, 15 pg/kg or less, 10 pg/kg or less, 5 pg/kg or less, 2.5 pg/kg or less, 2 pg/kg or less, 1.5 pg/kg or less, 1 pg/kg or less, 0.5 pg/kg or less, or 0.2 pg/kg or less of the anti-CD3 antibody such as
  • the dose on day 1 of the regimen is about 5-150 pg/m 2 /day, preferably about 55-150 pg/m 2 /day, for example about 60-100 pg/m 2 /day and escalates to the daily dose as recited immediately above by day 3, 4, 5, 6 or 7.
  • the subject on day 1, is administered a dose of approximately 60 pg/m 2 /day, on day 2 approximately 125 pg/m 2 /day, on day 3 approximately 250 pg/m 2 /day, on day 4 approximately 500 pg/m 2 /day and on subsequent days of the regimen (e.g., days 5-14) 1,000 pg/m 2 /day.
  • the subject on day 1, is administered a dose of approximately 60 pg/m 2 /day, on day 2 approximately 125 pg/m 2 /day, on day 3 approximately 250 pg/m 2 /day, on day 4 approximately 500 pg/m 2 /day and on subsequent days of the regimen (e.g., days 5-14) 1,030 pg/m 2 /day.
  • the subject on day 1, is administered a dose of approximately 100 pg/m 2 /day, on day 2 approximately 425 pg/m 2 /day, on day 3 approximately 850 pg/m 2 /day, on day 4 approximately 850 ug/m 2 /day and on subsequent days of the regimen (e.g., days 5-14) 1,000 pg/m 2 /day.
  • the subject on day 1, is administered a dose of approximately 60 pg/m 2 /day, on day 2 approximately 125 pg/m 2 /day, on day 3 approximately 250 pg/m 2 /day, on day 4 approximately 500 pg/m 2 /day and on subsequent days of the regimen (e.g., days 5-14) 1,070 pg/m 2 /day.
  • the initial dose is 1/4, to 1/2, to equal to the daily dose at the end of the regimen but is administered in portions at intervals of 6, 8, 10 or 12 hours.
  • a 13 pg/kg/day dose is administered in four doses of 3-4 pg/kg at intervals of 6 hours to reduce the level of cytokine release caused by administration of the antibody.
  • the first 1, 2, 3, or 4 doses or all the doses in the regimen are administered more slowly by intravenous administration.
  • a dose of 51 pg/m 2 /day may be administered over about 5 minutes, about 15 minutes, about 30 minutes, about 45 minutes, about 1 hour, about 2 hours, about 4 hours, about 6 hours, about 8 hours, about 10 hours, about 12 hours, about 14 hours, about 16 hours, about 18 hours, about 20 hours, and about 22 hours.
  • the dose is administered by slow infusion over a period of, e.g., 20 to 24 hours.
  • the dose is infused in a pump, preferably increasing the concentration of antibody administered as the infusion progresses.
  • a set fraction of the doses for, for example, the 60 pg nr/day to 1000 pg/nr/dav or the 65 pg m 2 /day to 1030 pg/m 2 /day regimen described above is administered in escalating doses.
  • the fraction is 1/10, 1/4, 1/3, 1/2, 2/3 or 3/4 of the daily doses of the regimens described above.
  • the anti-CD3 antibody such as teplizumab, otelixizumab or foralumab
  • the infusion may be constant or may start out at a lower dosage for, for example, the first 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours or 8 hours of the infusion and then increase to a higher dosage thereafter.
  • a dose equal to the amount administered in the 5 to 20-day regimens set forth above (for example the 14-day regimen set forth above).
  • a cumulative dose of approximately or greater than 9500 pg/m 2 , 10000 pg/m 2 , 10500 pg/m 2 , 11000 pg/m 2 , 11500 pg/m 2 , 12000 pg/m 2 , 12500 pg/m 2 , 13000 pg/m 2 , 13500 pg/m 2 or 14000 pg/m 2 is administered by infusion to the subject in need thereof.
  • a cumulative dose of teplizumab of approximately or greater than 11,240 pg/m 2 is administered by infusion to the subject in need thereof over a period of at least 30 minutes.
  • the speed and duration of the infusion is designed to minimize the level of free anti- CD3 antibody such as teplizumab, otelixizumab or foralumab in the subject after administration.
  • the level of free anti-CD3 antibody such as teplizumab should not exceed 200 ng/ml free antibody.
  • the infusion is designed to achieve a combined T cell receptor coating and modulation of at least 50%, 60%, 70%, 80%, 90%, 95% or of 100%.
  • the subject in need thereof is administered prior to the administering of the course IV infusion (for example the 14-day s course IV infusion) on each of days 1-5 an effective amount of pain reliever (such as a nonsteroidal anti-inflammatory drug (NSAID), acetaminophen), an antihistamine, an antiemetic or a combination thereof.
  • pain reliever such as a nonsteroidal anti-inflammatory drug (NSAID), acetaminophen), an antihistamine, an antiemetic or a combination thereof.
  • the pain reliever (such as a nonsteroidal anti-inflammatory drug (NSAID), acetaminophen), an antihistamine, an antiemetic or a combination thereof can be administered on each of days 1-5, 1-6, 1-7, 1-8, 1-9, 1-10, 1-11, 1-12, 1-13, 1-14 or during the duration of the treatment with the anti-CD3 antibodies.
  • the NSAID, acetaminophen, antihistamine, antiemetic or combination thereof is administered orally.
  • the NSAID, acetaminophen, antihistamine, antiemetic or combination thereof is administered intravenously.
  • antipyretics, antihistamines and/or antiemetics are administered to the subject in need thereof to mitigate cytokine release syndrome.
  • the liver enzymes are monitored and treatment with the anti-CD3 antibodies is discontinued or paused in subjects developing elevated ALT or AST more than 5 times the upper limit of normal.
  • the anti-CD3 antibody such as teplizumab, otelixizumab or foralumab is administered chronically to treat, prevent, or slow or delay the onset or progression, or ameliorate, mitigate, relieve, lighten, postpone, stop or halt one or more symptoms of type 1 diabetes.
  • a low dose of the anti-CD3 antibody such as teplizumab is administered once a month, twice a month, three times per month, once a week or even more frequently either as an alternative to the 6 to I4-day dosage regimen discussed above or after administration of such a regimen to enhance or maintain its effect.
  • Such a low dose may be anywhere from about 1 pg/m 2 to about 100 pg/m 2 . such as approximately 5 pg/m 2 . 10 pg/m 2 , 15 pg/m 2 , 20 pg/m 2 , 25 pg/m 2 , 30 pg/m 2 , 35 pg/m 2 , 40 pg/m 2 , 45 pg/m 2 , 50 pg/m 2 , 55 pg/m 2 , 60 pg/m 2 , 65 pg/m 2 , 70 pg/m 2 , 75 pg/m 2 , 80 pg/m 2 , 85 pg/m 2 , 90 pg/m 2 , 95 pg/m 2 or 100 pg/m 2 .
  • the anti-CD3 antibody such as teplizumab, otelixizumab or foralumab is administered by infusion in medical facilities, outpatient infusion centers.
  • the anti-CD3 antibody such as teplizumab, otelixizumab or foralumab is administered by infusion in a home setting.
  • Home infusion therapy involves the administration of the therapeutic agents, for example, anti-CD3 antibody using intravenous, or subcutaneous routes, in the patient's home rather than in a physician's office or hospital.
  • Infusion therapies in the home can be administered by a home health care worker or by a patient himself.
  • a health care worker having some training in the operation of infusion equipment and the administration of anti-CD3 antibody can provide the patient with self-administration training and all the necessary equipment and/or supplies needed for the administration.
  • the subject may be re-dosed at some time subsequent to administration of the anti-CD3 antibody such as teplizumab, otelixizumab or foralumab dosing regimen, for example, based upon one or more physiological parameters or may be done as a mater of course.
  • the anti-CD3 antibody such as teplizumab, otelixizumab or foralumab dosing regimen
  • Such redosing may be administered and/or the need for such redosing evaluated 2 months, 4 months, 6 months, 8 months, 9 months, 1 year, 15 months, 18 months, 2 years, 30 months or 3 years after administration of a dosing regimen and may include administering a course of treatment every 6 months, 9 months, 1 year, 15 months, 18 months, 2 years, 30 months or 3 years indefinitely.
  • Some embodiments relate to an anti-CD3 antibody for use in a method of preventing or delaying the onset of clinical type 1 diabetes (T1D), comprising administering a prophy lactically effective amount of an anti-CD3 antibody to a non-diabetic subject who is at risk of T1D, wherein the prophylactically effective amount has a cumulative dose of about 10,500 pg/m 2 to about 14,000 pg/m 2 ; and determining, prior to or after the administering step, that the non-diabetic subject has more than about 10% TIGIT+KLRG1+CD8+ T-cells in all CD3+ T cells, which is indicative of successful prevention or delay of the onset of clinical T1D.
  • T1D clinical type 1 diabetes
  • aspects of the disclosure relate to a method of preventing or delaying onset of clinical type 1 diabetes (T1D), comprising administering a prophylactically effective amount of an anti- CD3 antibody to a non-diabetic subject who is at risk of T1D, wherein the prophylactically effective amount has a cumulative dose of from about 10,500 pg/m 2 to about 14,000 pg/m 2 .
  • the subject is 8 years of age or older. In some embodiments, the subject is 5 years of age or older. In some embodiments, the subject is 3 years of age or older. In some embodiments, the subject is 1 year of age or older. In some embodiments, the subject is 1 year old or younger.
  • the non-diabetic subject is a relative of a patient with T1D.
  • the method further comprises determining that the non-diabetic subject (1) is negative for zinc transporter 8 (ZnT8) antibodies, (2) is HLA-DR4+, and/or (3) is not HLA-DR3+. In some embodiments, the non-diabetic subject does not have antibodies against ZnT8. In some embodiments, the non-diabetic subject is HLA-DR4+ and is not HLA-DR3+.
  • ZnT8 zinc transporter 8
  • the non-diabetic subject has 2 or more diabetes-related autoantibodies selected from islet cell antibodies (ICA), insulin autoantibodies (IAA), and antibodies to glutamic acid decarboxylase (GAD), tyrosine phosphatase (IA-2/ICA512) or ZnT8.
  • ICA islet cell antibodies
  • IAA insulin autoantibodies
  • GAD glutamic acid decarboxylase
  • IA-2/ICA512 tyrosine phosphatase
  • ZnT8 ZnT8.
  • the non-diabetic subject has abnormal glucose tolerance on oral glucose tolerance test (OGTT).
  • the abnormal glucose tolerance on OGTT is a fasting glucose level of 110-125 mg/dL, or 2 hour plasma of > 140 and ⁇ 200 mg/dL, or an intervening glucose value at 30, 60, or 90 minutes on OGTT > 200 mg/dL.
  • the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab. In some embodiments, the anti-CD3 antibody is teplizumab.
  • the prophy tactically effective amount comprises a 14-day course of subcutaneous (SC)inj ection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody with a cumulative dose of from about 11,000 pg/m 2 to about 14,000 pg/m 2 .
  • the method comprises administering a 14-day course IV infusion at about 60 pg/m 2 on day 1, about 125 pg/m 2 on day 2, about 250 pg/m 2 on day 3, and about 500 pg/m 2 on day 4, and a dose of about 1,000 pg/m 2 on each of days 5-14.
  • the method comprises administering a 14-day course IV infusion at about 60 pg/m 2 , about 125 pg/m 2 on day 1, about 250 pg/m 2 on day 2, and about 500 pg/m 2 on day 3, and a dose of about 1,030 pg/m 2 on each of days 5-14.
  • the method comprises administering a 14-day course IV infusion at about 100 pg/m 2 on day 1, about 425 pg/m 2 on day 2, about 850 pg/m 2 on day 3, about 850 pg/m 2 on day 4, and a dose of about 1,000 pg/m 2 on each of days 5-14.
  • the method comprises administering a 14-day course IV infusion at about 65 pg/m 2 on day 1, about 125 pg/m 2 on day 2, about 250 pg/m 2 on day 3, and about 500 pg/m 2 on day 4, and a dose of about 1,070 pg/m 2 on each of days 5-14.
  • the method comprises administering a 14-day course IV infusion at about 65 pg/m 2 on day 1, about 125 pg/m 2 on day 2, about 250 pg/m 2 on day 3, and about 500 pg/m 2 on day 4, and a dose of about 1,030 pg/m 2 on each of days 5-14.
  • the prophylactically effective amount delays median time to clinical diagnosis of T1D by at least 50%, at least 80%, or at least 90%. In some embodiments, the prophylactically effective amount delays median time to clinical diagnosis of T1D by at least 12 months, at least 18 months, at least 24 months, at least 36 months, at least 48 months, or at least 60 months.
  • the method further comprises determining, prior to or after the administering step, that the non-diabetic subject has more than about 10% TIGIT+KLRG1+CD8+ T-cells in all CD3+ T cells, which is indicative of successful prevention or delay of the onset of clinical T1D.
  • the determining of TIGIT+KLRG1+CD8+ T-cells is by flow cytometry.
  • the method further comprises determining a decrease in a percentage of CD8+ T cells expressing proliferation markers Ki67 and/or CD57.
  • aspects of the disclosure relate to a method of preventing or delaying onset of clinical type 1 diabetes (T1D), comprising administering to a non-diabetic subject 8 years of age or older who is at risk of T1D a 14-day course IV infusion of teplizumab at about 65 pg/m 2 on day 1, about 125 pg/m 2 on day 2, about 250 pg/m 2 on day 3, and about 500 pg/m 2 on day 4, and a dose of about 1,030 pg/m 2 on each of days 5-14.
  • T1D clinical type 1 diabetes
  • aspects of the disclosure relate to a method of delaying onset of Stage 3 type 1 diabetes (T1D), comprising administering to a subject in need thereof who has a diagnosis of stage 2 T1D a 14-day course IV infusion of teplizumab at about 65 pg/m 2 on day 1, about 125 pg/m 2 on day 2, about 250 pg/m 2 on day 3, and about 500 pg/m 2 on day 4, and a dose of about 1,030 pg/m 2 on each of days 5-14.
  • T1D Stage 3 type 1 diabetes
  • the subject in need thereof is an adult. In some embodiments, the subject in need thereof is a pediatric subject 8 years of age or older. In some embodiments, the subject in need thereof is a pediatric subject 5 years of age or older. In some embodiments, the subject in need thereof is a pediatric subject 3 years of age or older. In some embodiments, the subject in need thereof is a pediatric subject 1 year of age or older. In some embodiments, the subject in need thereof is a pediatric subject 1 year old or younger.
  • the method comprises documenting at least two positive pancreatic islet autoantibodies in the subject who has dysglycemia without overt hyperglycemia before administering the 14-day course.
  • the subject in need thereof has dysglycemia without overt hyperglycemia and has two or more pancreatic islet autoantibodies.
  • the two or more pancreatic islet autoantibodies comprise islet cell antibodies (ICA), insulin autoantibodies (IAA), and antibodies to glutamic acid decarboxylase (GAD), tyrosine phosphatase (IA-2/ICA512) or ZnT8.
  • the method comprises administering on at least each of days 1-5, and prior to the administering of the 14- day course IV infusion, an effective amount of a nonsteroidal anti-inflammatory drug (NSAID), acetaminophen, an antihistamine, an anti emetic or a combination thereof.
  • NSAID nonsteroidal anti-inflammatory drug
  • the method comprises administering orally the NSAID, acetaminophen, antihistamine, antiemetic or combination thereof.
  • the method comprises administering the 14-day course IV infusion once daily for 14 consecutive days over a period of at least 30 minutes.
  • aspects of the disclosure relate to a method of prognosing responsiveness of an anti- CD3 antibody in preventing or delaying onset of type 1 diabetes (T1D), the method comprising administering a prophylactically effective amount of an anti-CD3 antibody to a non-diabetic subject who is at risk of T1D, wherein the prophylactically effective amount has a cumulative dose of about 10,500 pg/m 2 to about 14,000 pg/m 2 ; and determining C-peptide area under the curve (AUC): glucose AUC ratio, wherein an increase in the ratio indicates responsiveness to the anti-CD3 antibody and/or non-progression to clinical T1D.
  • AUC C-peptide area under the curve
  • aspects of the disclosure relate to an anti-CD3 antibody for use in a method of preventing or delaying onset of clinical type 1 diabetes (T1D), comprising: administering a prophylactically effective amount of an anti-CD3 antibody to a non-diabetic subject who is at risk of T1D, wherein the prophylactically effective amount has a cumulative dose of about 10,500 pg/m 2 to about 14,000 pg/m 2 .
  • a teplizumab composition for use in a method of delaying onset of Stage 3 type 1 diabetes (T1D), comprising administering to a subject in need thereof who has a diagnosis of stage 2 T1D a 14-day course IV infusion at about 65 pg/m 2 on day 1, about 125 pg/m 2 on day 2, about 250 pg/m 2 on day 3, and about 500 pg/m 2 on day 4, and a dose of about 1,030 pg/m 2 on each of days 5-14.
  • T1D Stage 3 type 1 diabetes
  • Type 1 diabetes is an autoimmune disease characterized by T-cell mediated destruction of insulin producing beta cells within the pancreatic islets of Langerhans. Longitudinal observational studies over more than 30 years have described the progression of the autoimmune disease from the first appearance of autoantibodies until beta cell function is critically impaired and the clinical diagnosis, often with ketoacidosis, occurs (7-5). T1D is associated with a need for lifelong exogenous insulin administration for survival, increased morbidity and mortality due to immediate (e.g. hypoglycemia) and long-term complications (e.g. vascular, renal, and eye disease), and reduced life-span, life impairments, and considerable health-care-related costs (6-9). Thus, approaches to prevent progression to clinical T1D before irremediable beta cell destruction and insulin deficiency, are of paramount importance.
  • beta cell function may fluctuate between abnormal and normal values within an individual who is at risk (15, 16).
  • OGTT glucose tolerance classifications used to designate a clinical diagnosis, and beta cell function, measured by C-peptide responses to a metabolic challenge may not be closely related and at diagnosis using an OGTT, many individuals have clinically meaningful C-peptide responses (15-18).
  • dosing regimens matching AUCinf, Cmax and Ctroughis (trough concentration prior to Day 14 dosing) exposure parameters of the reference regimen Herold 14- day regimen used in Protege and TN- 10 studies, Herold et al NEJM 2019).
  • Each of these regimens includes a 4-day ramp-up followed by 10 consecutive days of maintenance dosing, with an infusion duration of 30 minutes.
  • the ramp-up is important to avoid the occurrence of acute events such as those related to cytokine release.
  • Regimen A Incorporates the backbone of the TN-10 (Herold 14-day) 4-day ramp-up and maintenance doses, with an approximate 18-20% upward adjustment for each daily dose.
  • This regimen meets the bioequivalence criteria for the three PK parameters under both conditional and typical (average) simulations with the exception that the average (typical) simulation ratio for CtroughB is slightly less than 0.80 (0.779). (Table 2).
  • the cumulative dose for this regimen is 10,935 pg/m 2 (approximately 21% higher compared to the Herold 14-day regimen).
  • the cumulative dose for this regimen is 11,235 pg/m 2 (approximately 24% higher compared to the Herold 14-day regimen).
  • o Day 1 60 pg/m 2
  • o Day 2 125 pg/m 2
  • Day 3 250 pg/m 2
  • Day 4 500 pg/m 2 o
  • Days 5-14 1030 pg/m 2
  • Regimen C Incorporates the backbone of the PROTECT 12-day regimen with a 2-day ramp- up, with minor adjustment for the Day 1 dose from 106 pg/m 2 to 100 pg/m 2 to simplify dose calculations and preparations; the two doses of 850 pg/m 2 on Day 3 and Day 4 replicate the PROTECT regimen before the administration of the maintenance dose of 1000 pg/m 2 .
  • This regimen meets 80-125% bioequivalence criteria for AUCinf, Cmax and Ctroughia under both the conditional and typical (average) simulations (Table 2).
  • the cumulative dose for this regimen is 12,225 pg/m 2 (approximately 35% higher compared to the Herold 14-day regimen).
  • FIG. 1 and FIG. 5 A comparison of the distribution of exposures (AUCinf, Cmax and Ctroughts) for the reference regimen (Herold 14-day) and the anti-CD3 antibody exposures predicted for the alternative regimens are shown in FIG. 1 and FIG. 5.
  • FIG. 2 and FIG. 6 A comparison of the distribution of exposures (AUCinf, Cmax and Ctroughis) for the Protege and TN-10 regimen (Herold 14-day) and the anti-CD3 antibody exposures predicted for the alternative regimens are shown in FIG. 2 and FIG. 6.
  • Example 4 Dosage and Administration [00165] The teplizumab compositions and methods provided herein are indicated to delay the onset of Stage 3 type 1 diabetes in adults and pediatric patients 8 years of age and older with Stage 2 type 1 diabetes.
  • Teplizumab-mzwv (also referred herein as Teplizumab) is a CD3-directed monoclonal antibody (humanized IgGl kappa) that has a molecular weight of approximately 150 kilodalton (kDa) and is expressed from a recombinant Chinese hamster ovary (CHO) cell line.
  • Teplizumab injection is supplied as a sterile, preservative-free, clear and colorless solution in a 2 mg/2 mL (1 mg/mL) single-dose vial for intravenous use.
  • Each mL contains 1 mg of teplizumab-mzwv, dibasic sodium phosphate (0.26 mg), monobasic sodium phosphate (0.98 mg), polysorbate 80 (0.05 mg), sodium chloride (8.78 mg), and water for injection.
  • the pH is 6.1.
  • Teplizumab-mzwv binds to CD3 (a cell surface antigen present on T lymphocytes) and delays the onset of Stage 3 type 1 diabetes in adults and pediatric patients aged 8 years and older with Stage 2 type 1 diabetes.
  • the mechanism may involve partial agonistic signaling and deactivation of pancreatic beta cell auto-reactive T lymphocytes.
  • Teplizumab-mzwv leads to an increase in the proportion of regulatory T cells and of exhausted CD8+ T cells in peripheral blood.
  • teplizumab-mzwv binds to CD3 molecules on the surface of both CD4+ and CD8+ T cells during treatment, with internalization of the teplizumab- mzwv/CD3 complex from the surface of T cells.
  • Pharmacodynamic effects include lymphopenia in the absence of depletion of T cells with a nadir on the 5th day of dosing, during a 14-day course of teplizumab-mzwv treatment.
  • Teplizumab-mzwv exposure-response relationship and time course of pharmacodynamic response for the safety and effectiveness of teplizumab-mzwv have not been fully characterized.
  • Teplizumab-mzwv showed saturable binding and elimination.
  • the mean (SD) terminal elimination half-life and clearance of teplizumab-mzwv are 4.5 (0.2) days and 2.7 (0.8) L/day in a 60 kg subject, respectively.
  • Teplizumab-mzwv is expected to be metabolized into small peptides by catabolic pathways.
  • BSA-based dosing normalizes the exposure to teplizumab-mzwv across body weight.
  • Each vial is intended for single-dose only.
  • PVC Polyvinylchloride
  • Premedicate prior to infusion for the first 5 days of dosing with oral medications including: (1) a nonsteroidal anti-inflammatory drug (NSAID) or acetaminophen, (2) an antihistamine, and/or (3) an antiemetic. Administer additional doses of premedication if needed.
  • NSAID nonsteroidal anti-inflammatory drug
  • acetaminophen an antihistamine
  • an antiemetic an antiemetic
  • Epstein-Barr virus EBV
  • CMV cytomegalovirus
  • Teplizumab preserves C-peptide in recent-onset type 1 diabetes: two-year results from the randomized, placebo- controlled Protege trial. Diabetes 62, 3901 -3908 (2013).

Landscapes

  • Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Diabetes (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Endocrinology (AREA)
  • Hematology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Emergency Medicine (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Obesity (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Provided herein, in one aspect, is a method of preventing or delaying the onset of clinical type 1 diabetes (T1D), comprising: administering a prophylactically effective amount of an anti-CD3 antibody to a non-diabetic subject who is at risk of T1D, wherein the prophylactically effective amount has a cumulative dose of about 10,500 μg/m2 to about 14,000 μg/m2.

Description

METHODS AND COMPOSITIONS FOR PREVENTING OR DELAYING TYPE 1 DIABETES
RELATED APPLICATIONS
[0001] This application claims the benefit of and priority to U.S. Provisional Patent Application Serial No. 63/345,365. filed May 24, 2022, U.S. Provisional Patent Application Serial No. 63/367,992, filed July 8, 2022, U.S. Provisional Patent Application Serial No. 63/382,382, filed November 4, 2022, U.S. Utility Patent Application No. 18/321,964, filed May 23, 2023, and Taiwanese Patent Application No. 112119143, filed May 23, 2023, the entire contents of each of which are hereby incorporated by reference in their entireties.
SEQUENCE LISTING
[0002] This specification includes a sequence listing submitted herewith, which includes the file entitled 178833-011704. xml having the following size: 4,136 bytes which was created May 8, 2023, the contents of which are incorporated by reference herein.
FIELD
[0003] The present disclosure relates in general to compositions and methods of preventing or delaying the onset of clinical type 1 diabetes (T1D) in subjects at risk, and more particularly the use of anti-CD3 antibodies.
BACKGROUND
[0004] Type 1 diabetes (T1D) is caused by the autoimmune destruction of insulin producing beta cells in the islets of Langerhans leading to dependence on exogeneous insulin injections for survival. Approximately 1.6 million Americans have Type 1 diabetes, and after asthma, it remains one of the most common diseases of childhood. Despite improvements in care, most affected individuals with T1D are not able to consistently achieve desired glycemic targets. For individuals with type 1 diabetes, there are persisting concerns for increased risk of both morbidity and mortality. Two recent studies noted loss of 17.7 life-years for children diagnosed before age 10, and 11 and 13 life-years lost for adult-diagnosed Scottish men and women respectively.
[0005] In genetically susceptible individuals, T1D progresses through asymptomatic stages prior to overt hyperglycemia, characterized first by the appearance of autoantibodies (Stage 1) and then dysglycemia (Stage 2). In Stage 2, metabolic responses to a glucose load are impaired but other metabolic indices, for example glycosylated hemoglobin, are normal and insulin treatment is not needed. These immunologic and metabolic features identify individuals who are at high-risk for development of clinical disease with overt hyperglycemia and requirement for insulin treatment (Stage 3). Several immune interventions have been shown to delay decline in beta cell function when studied in recent-onset clinical T1D. One promising therapy is the FcR non-binding anti-CD3 monoclonal antibody teplizumab, as several studies have shown that short-term treatment reduces loss of P cell function durably, with an observable effect seen as long as 7 years after diagnosis and treatment. The drug modifies the function of CD8+ T lymphocytes, which are thought to be important effector cells that cause beta cell killing.
[0006] To date, no intervention initiated before the clinical diagnosis (i.e., at Stage 1 or 2) has altered progression to clinical, Stage 3 T1D. Thus, a need exists for a treatment that would prevent or delay the onset of clinical T1D in high-risk individuals.
SUMMARY
[0007] Aspects of the disclosure relate to a method of preventing or delaying onset of clinical type 1 diabetes (T1D), comprising: administering a prophylactically effective amount of an anti- CD3 antibody to a non-diabetic subject who is at risk for T1D.
[0008] In some embodiments, the method of preventing or delaying onset of clinical type 1 diabetes (T1D), comprises administering a prophylactically effective amount of an anti-CD3 antibody to a non-diabetic subject who is at risk of T1D, wherein the prophylactically effective amount has a cumulative dose of from about 10,500 pg/m2 to about 14,000 pg/m2.
[0009] In some embodiments, the non-diabetic subject is an adult or a pediatric subject. In some embodiments, the pediatric subject is 8 years old or older. In some embodiments, the pediatric subject is 7 years old or older. In some embodiments, the pediatric subject is 6 years old or older. In some embodiments, the pediatric subject is 5 years old or older. In some embodiments, the pediatric subject is 4 years old or older. In some embodiments, the pediatric subject is 3 years old or older. In some embodiments, the pediatric subject is 3 years old or older. In some embodiments, the pediatric subject is 2 year old or older. In some embodiments, the pediatric subject is 1 year old or older. In some embodiments, the pediatric subject is 1 year old or younger. In some embodiments, the pediatric subject is an infant.
[0010] In some embodiments, the non-diabetic subject is a relative of a patient with T1D. [0011] In some embodiments, the method further includes determining that the non-diabetic subject (1) is negative for zinc transporter 8 (ZnT8) antibodies, (2) is HLA-DR4+, and/or (3) is not HLA-DR3+.
[0012] In some embodiments, the non-diabetic subject has 2 or more diabetes-related autoantibodies selected from islet cell antibodies (ICA), insulin autoantibodies (IAA), and antibodies to glutamic acid decarboxylase (GAD), ty rosine phosphatase (IA-2/ICA512) or ZnT8.
[0013] In some embodiments, the non-diabetic subject has abnormal glucose tolerance on oral glucose tolerance test (OGTT). In some embodiments, the abnormal glucose tolerance on OGTT is a fasting glucose level of 110-125 mg/dL, or 2 hour plasma of > 140 and < 200 mg/dL, or an intervening glucose value at 30, 60, or 90 minutes on OGTT > 200 mg/dL.
[0014] In some embodiments, the non-diabetic subject has abnormal results in a glucose monitoring system (CGM) revealing high sensor average glucose levels (>=110 mg/dL), or high vanabihty of glycemia (CV >=15), or less time in range (>=10% of the time above 140 mg/dL).
[0015] In some embodiments, the non-diabetic subject does not have antibodies against ZnT8.
[0016] In some embodiments, the non-diabetic subject is HLA-DR4+ and is not HLA-DR3+.
[0017] In some embodiments, the non-diabetic subject does not have antibodies against ZnT8, is HLA-DR4+ and is not HLA-DR3+.
[0018] In some embodiments, the non-diabetic subject is a relative of a patient with T1D and does not have antibodies against ZnT8. In some embodiments, the non-diabetic subject is a relative of a patient with T1D, does not have antibodies against ZnT8, is HLA-DR4+ and is not HLA-DR3+.
[0019] In some embodiments, the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab. In some embodiments, the anti-CD3 antibody is teplizumab.
[0020] In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14-day course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at a cumulative dose greater than 10,500 micrograms/meter squared (pg/m2). In some embodiments, the prophylactically effective amount of the antibody comprises a 14-day course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at a cumulative dose of from about 11,000 pg/m2 to about 14,000 pg/m2. In some embodiments, the anti-CD3 antibody is or comprises teplizumab.
[0021] In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14 day course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 10,500 to about 14,000 pg/m2, from about
10.500 to about 13,500 pg/m2, from about 10,500 to about 13,000 pg/m2, from about 10,500 to about 12,500 pg/m2, from about 10,500 to about 12,000 pg/m2, from about 10,500 to about
11.500 pg/m2, or from about 10,500 to about 11,000 pg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14-day course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at about 10,500 pg/m2, 11,000 pg/m2, 11,500 pg/m2, 12,000 pg/m2, 12,500 pg/m2, 13,000 pg/m2, 13,500 pg/m2, or 14,000 pg/m2. In some embodiments, the anti-CD3 antibody is or comprises teplizumab.
[0022] In some embodiments, the method comprises administering to the non-diabetic subj ect who is at risk for T1D a 14-day course IV infusion of the anti-CD3 antibody at about 60 pg/m2, about 125 pg/m2, about 250 pg/m2, and about 500 pg/m2, on days 1-4, respectively, and a dose of about 1,000 pg/m2 on each of days 5-14. In some embodiments, the cumulative dose is about 10,935 pg/m2. In some embodiments, the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab. In some embodiments, the anti-CD3 antibody is teplizumab. In some embodiments, the method comprises administering to the non-diabetic subject who is at risk for T1 D a 14-day course IV infusion of teplizumab at about 60 pg/m2, about 125 pg/m2, about 250 pg/m2, and about 500 pg/m2, on days 1-4, respectively, and a dose of about 1,000 pg/m2 on each of days 5-14.
[0023] In some embodiments, the method comprises administering to the non-diabetic subj ect who is at risk for T1D a 14-day course IV infusion of the anti-CD3 antibody at about 60 pg/m2, about 125 pg/m2, about 250 pg/m2, and about 500 pg/m2, on days 1-4, respectively, and a dose of about 1,030 pg/m2 on each of days 5-14. In some embodiments, the cumulative dose is about 11,235 pg/m2. In some embodiments, the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab. In some embodiments, the anti-CD3 antibody is teplizumab. In some embodiments, the method comprises administering to the non-diabetic subject who is at risk for T1D a 14-day course IV infusion of teplizumab at about 60 pg/m2, about 125 pg/m2, about 250 pg/m2, and about 500 pg/m2, on days 1-4, respectively, and a dose of about 1,030 pg/m2 on each of days 5-14.
[0024] In some embodiments, the method comprises administering to the non-diabetic subj ect who is at risk for T1D a 14-day course IV infusion of the anti-CD3 antibody at about 100 pg/m2, about 425 pg/m2, about 850 pg/m2, and about 850 pg/m2, on days 1-4, respectively, and a dose of about 1,000 pg/m2 on each of days 5-14. In some embodiments, the cumulative dose is about 12,225 pg/m2. In some embodiments, the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab. In some embodiments, the anti-CD3 antibody is teplizumab. In some embodiments, the method comprises administering to the non-diabetic subject who is at risk for T1D a 14-day course IV infusion of teplizumab at about 100 pg/m2, about 425 pg/m2, about 850 pg/m2, and about 850 pg/m2, on days 1-4, respectively, and a dose of about 1,000 pg/m2 on each of days 5-14.
[0025] In some embodiments, the method comprises administering to the non-diabetic subj ect who is at risk for T1D a 14-day course IV infusion of the anti-CD3 antibody at about 65 pg/m2, about 125 pg/m2, about 250 pg/m2, and about 500 pg/m2, on days 1-4, respectively, and a dose of about 1,070 pg/m2 on each of days 5-14. In some embodiments, the cumulative dose is about 11,640 pg/m2. In some embodiments, the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab. In some embodiments, the anti-CD3 antibody is teplizumab. In some embodiments, the method comprises administering to the non-diabetic subject who is at risk for T1D a 14-day course IV infusion of teplizumab at about 65 pg/m2, about 125 pg/m2, about 250 pg/m2, and about 500 pg/m2, on days 1-4, respectively, and a dose of about 1,070 pg/m2 on each of days 5-14.
[0026] In some embodiments, the method comprises administering to the non-diabetic subj ect who is at risk for T1D a 14-day course IV infusion of the anti-CD3 antibody at about 65 pg/m2, about 125 pg/m2, about 250 pg/m2, and about 500 pg/m2, on days 1-4, respectively, and a dose of about 1,030 pg/m2 on each of days 5-14. In some embodiments, the cumulative dose is about 11,240 pg/m2. In some embodiments, the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab. In some embodiments, the anti-CD3 antibody is teplizumab. In some embodiments, the method comprises administering to the non-diabetic subject who is at risk for T1D a 14-day course IV infusion of teplizumab at about 65 pg/m2, about 125 pg/m2, about 250 pg/m2, and about 500 pg/m2, on days 1-4, respectively, and a dose of about 1,030 pg/m2 on each of days 5-14. [0027] In some embodiments, the prophylactically effective amount delays median time to clinical diagnosis of T1D by at least 50%, at least 80%, or at least 90%. In some embodiments, the prophylactically effective amount delays median time to clinical diagnosis of T1D by at least 12 months, at least 18 months, at least 24 months, at least 36 months, at least 48 months, or at least 60 months.
[0028] In some embodiments, the method further comprises determining, prior to or after the administering step, that the non-diabetic subject has more than about 10% TIGIT+KLRG1+CD8+ T-cells in all CD3+ T cells, which is indicative of successful prevention or delay of the onset of clinical T1D. In some embodiments, the step of determining of TIGIT+KLRG1+CD8+ T-cells is by flow cytometry. In some embodiments, the method further comprises determining a decrease in a percentage of CD8+ T cells expressing proliferation markers Ki67 and/or CD57.
[0029] Some aspects of the disclosure relate to a method of preventing or delaying onset of clinical type 1 diabetes (T1D), comprising: administering to a non-diabetic subject 8 years of age or older who is at risk of T1D a 14 day course IV infusion of teplizumab at about 65 pg/m2 on day 1, about 125 pg/m2 on day 2, about 250 pg/m2 on day 3, and about 500 pg/m2 on day 4, and a dose of about 1,030 pg/m2 on each of days 5-14.
[0030] Some aspects of the disclosure relate to a method of delaying onset of Stage 3 type 1 diabetes (T1D), comprising administering to a subject in need thereof who has a diagnosis of stage 2 T1D a 14 day course IV infusion of teplizumab at about 65 pg/m2 on day 1, about 125 pg/m2 on day 2, about 250 pg/m2 on day 3, and about 500 pg/m2 on day 4, and a dose of about 1,030 pg/m2 on each of days 5-14. In some embodiments, the non-diabetic subject is an adult or a pediatric subject. In some embodiments, the pediatric subject is 8 years old or older. In some embodiments, the pediatric subject is 7 years old or older. In some embodiments, the pediatric subject is 6 years old or older. In some embodiments, the pediatric subject is 5 years old or older. In some embodiments, the pediatric subject is 4 years old or older. In some embodiments, the pediatric subject is 3 years old or older. In some embodiments, the pediatric subject is 3 years old or older. In some embodiments, the pediatric subject is 2 year old or older. In some embodiments, the pediatric subject is 1 year old or older. In some embodiments, the pediatric subject is 1 year old or younger. In some embodiments, the pediatric subject is an infant. [0031] In some embodiments, the method comprises documenting at least two positive pancreatic islet autoantibodies in the subject who has dysglycemia without overt hyperglycemia before administering the 14 day course.
[0032] In some embodiments, the subj ect in need thereof has dysglycemia without overt hyperglycemia and has two or more pancreatic islet autoantibodies.
[0033] In some embodiments, the two or more pancreatic islet autoantibodies comprise islet cell antibodies (ICA), insulin autoantibodies (IAA), and antibodies to glutamic acid decarboxylase (GAD), tyrosine phosphatase (IA-2/ICA512) or ZnT8.
[0034] In some embodiments, the method comprises administering on at least each of days 1- 5, and prior to the administering of the 14 days course IV infusion, an effective amount of a nonsteroidal anti-inflammatory drug (NSAID), acetaminophen, an antihistamine, an antiemetic or a combination thereof. In some embodiments, the method comprises administering orally the NSAID, acetaminophen, antihistamine, antiemetic or combination thereof.
[0035] In some embodiments, the method comprises administering the 14 days course IV infusion once daily for 14 consecutive days over a period of at least 30 minutes.
[0036] In some embodiments, the method further comprises determining, pnor to or after the administering step, that the non-diabetic subject has more than about 10% TIGIT+KLRG1+CD8+ T-cells in all CD3+ T cells, which is indicative of successful prevention or delay of the onset of clinical T1D.
[0037] In some embodiments, the determining of TIGIT+KLRG1+CD8+ T-cells is by flow cytometry.
[0038] In some embodiments, the method further includes determining a decrease in a percentage of CD8+ T cells expressing proliferation markers Ki67 and/or CD57.
[0039] Other aspects of the disclosure relate to a method of prognosing responsiveness of an anti-CD3 antibody in preventing or delaying onset of type I diabetes (T1D), the method comprising administering a prophy tactically effective amount of an anti-CD3 antibody to a nondiabetic subject who is at risk for T1D, wherein the prophylactically effective amount has a cumulative dose of about 10,500 pg/rrr to about 14,000 pg/m2; and determining C-peptide area under the curve (AUC): glucose AUC ratio, wherein an increase in the ratio indicates responsiveness to the anti-CD3 antibody and/or non-progression to clinical T1D. [0040] Aspects of the disclosure relate to an anti-CD3 antibody for use in a method of preventing or delaying onset of clinical type 1 diabetes (T1D), comprising administering a prophylactically effective amount of an anti-CD3 antibody to a non-diabetic subject who is at risk of T1D, wherein the prophylactically effective amount has a cumulative dose of about 10,500 pg/m2 to about 14,000 pg/m2. In some embodiments, the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab. In some embodiments, the anti-CD3 antibody is teplizumab.
[0041] Aspects of the disclosure relate to a method of preventing or delaying onset of Stage 3 type 1 diabetes (T1D), comprising administering to a subject who has a diagnosis of stage 2 T1D a 14 day course IV infusion of teplizumab at about 65 pg/m2 on day 1, about 125 pg/m2 on day 2, about 250 pg/m2 on day 3, and about 500 pg/m2 on day 4, and a dose of about 1,030 pg/m2 on each of days 5-14.
[0042] In some embodiments, the subject in need thereof has two or more pancreatic islet autoantibodies. In some embodiments, the subject in need thereof has dysglycemia without overt hyperglycemia and has two or more pancreatic islet autoantibodies. In some embodiments, the method comprises confirming stage 2 T1D by documenting at least two positive pancreatic islet autoantibodies in subjects who have dysglycemia without overt hyperglycemia before administering the 14 day course. In some embodiments, the pancreatic islet autoantibodies comprise islet cell antibodies (ICA), insulin autoantibodies (IAA), antibodies to glutamic acid decarboxylase (GAD), tyrosine phosphatase (IA-2/ICA512) or ZnT8.
BRIEF DESCRIPTION OF THE DRAWINGS
[0043] FIG. 1 shows AUCinf, Cmax and Ctroughis: Reference Regimen and Simulated Alternative Regimens (Regimen A, Regimen B, regimen C). Exposures for the reference regimen reflect the Herold 14-day regimen administered to PROTECT study participants who received the anti- CD3 antibody. Exposures for alternative regimens reflect simulated anti-CD3 antibody administered to a similar population of T1D patients as a subset of PROTECT study participants. The boxes indicate the median (line within a box) values as well as QI to Q3 interquartile ranges (IQRs). The whiskers indicate Ql-1.5 IQR and Q3+1.5 IQR. The data points outside the whiskers indicate outlier values.
[0044] FIG. 2 shows AUCinf, Cmax and Ctroughis: Historical Clinical Trials and Simulated Alternative Regimens for the anti-CD3 antibody. Protege and TN-I0 exposures represent all patients who received the Herold 14-day regimen in these studies; PROTECT exposures represent patients who received the anti-CD3 antibody in the PROTECT study. Regimen A, Regimen B and Regimen C graphs are simulated PK data for anti-CD3 antibody Regimen A, Regimen B and Regimen C using in a similar population of T1D patients as a subset of PROTECT study subjects. The boxes indicate the median (line within a box) values as well as QI to Q3 interquartile ranges (IQRs). The whiskers indicate Ql-1.5 IQR and Q3+1.5 IQR. The data points outside the whiskers indicate outlier values.
[0045] FIG. 3 shows Predicted Cmax and Ctrough Levels During 14-day Treatment Course: Regimens A and B. The boxes indicate the median (line within a box) values as well as QI to Q3 interquartile ranges (IQRs). The whiskers indicate Ql-1.5 IQR and Q3+1.5 IQR. The data points outside the whiskers indicate outlier values.
[0046] FIG. 4 shows Predicted Cmax and Ctrough Levels During 14-day Treatment Course: Regimen C. The boxes indicate the median (line within a box) values as well as QI to Q3 interquartile ranges (IQRs). The whiskers indicate Ql-1.5 IQR and Q3+1.5 IQR. The data points outside the whiskers indicate outlier values.
[0047] FIG. 5 shows AUCinf, Cmax and Ctroughis: Reference Regimen and Simulated Regimen D. Exposures for the reference regimen reflect the Herold 14-day regimen administered to PROTECT study participants who received the anti-CD3 antibody. Exposures for alternative regimens reflect simulated anti-CD3 antibody administered to a similar population of T1D patients as a subset of PROTECT study participants. The boxes indicate the median (line within a box) values as well as QI to Q3 interquartile ranges (IQRs). The whiskers indicate Ql-1.5 IQR and Q3+1.5 IQR. The data points outside the whiskers indicate outlier values.
[0048] FIG. 6 shows AUCinf, Cmax and Ctmughis: Historical Clinical Trials and Simulated Regimen D for the anti-CD3 antibody. Protege and TN-10 exposures represent all patients who received the Herold 14-day regimen in these studies; PROTECT exposures represent patients who received the anti-CD3 antibody in the PROTECT study. Regimen D is simulated PK data for anti-CD3 antibody Regimen D using in a similar population of T1D patients as a subset of PROTECT study subjects. The boxes indicate the median (line within a box) values as well as QI to Q3 interquartile ranges (IQRs). The whiskers indicate Ql-1.5 IQR and Q3+1.5 IQR. The data points outside the whiskers indicate outlier values. [0049] FIG. 7 shows Predicted Cmax and Ctrough Levels During 14-day Treatment Course: Regimen D. The boxes indicate the median (line within a box) values as well as QI to Q3 interquartile ranges (IQRs). The whiskers indicate Ql-1.5 IQR and Q3+1.5 IQR. The data points outside the whiskers indicate outlier values.
DETAILED DESCRIPTION
[0050] Provided herein, in some embodiments, is a method of preventing or delaying the onset of clinical type 1 diabetes (T1D), comprising: providing a non-diabetic subject who is at risk for T1D; administering a prophylactically effective amount of an anti-CD3 antibody to the nondiabetic subject; and determining, prior to or after the administering step, that the non-diabetic subject has more than about 10% TIGIT+KLRG1+CD8+ T-cells in all CD3+ T cells, which is indicative of successful prevention or delay of the onset of clinical T1D.
[0051] In some embodiments, a method of prognosing responsiveness of an anti-CD3 antibody, e.g., tephzumab, in preventing or delaying the onset of T1D is provided. The method can include: providing a non-diabetic subject who is at risk for T1D; administering a prophylactically effective amount of the anti-CD3 antibody, e.g., tephzumab, to the non-diabetic subject; and determining C-peptide area under the curve (AUC): glucose AUC ratio, wherein an increase in the ratio indicates responsiveness to the anti-CD3 antibody.
Definitions
[0052] Certain terms are defined herein below. Additional definitions are provided throughout the application.
[0053] As used herein, the articles “a” and “an” refer to one or more than one, e.g., to at least one, of the grammatical object of the article. The use of the words "a" or "an" when used in conjunction with the term "comprising" herein may mean "one," but it is also consistent with the meaning of "one or more," "at least one," and "one or more than one."
[0054] As used herein, “about” and “approximately” generally mean an acceptable degree of error for the quantity measured given the nature or precision of the measurements. Exemplary degrees of error are within 30 percent (%), within 25% within 20 %, for example, within 10%, or, within 5% of a given range of values. The term “substantially” means more than 50%, more than 60%, more than 70% more than 80%, and more than 90% or 95%. [0055] As used herein the term "comprising" or "comprises" is used in reference to compositions, methods, and respective component(s) thereof, that are present in a given embodiment, yet open to the inclusion of unspecified elements.
[0056] As used herein the term "consisting essentially of refers to those elements required for a given embodiment. The term permits the presence of additional elements that do not materially affect the basic and novel or functional characteristic(s) of that embodiment of the disclosure.
[0057] The term "consisting of refers to compositions, methods, and respective components thereof as described herein, which are exclusive of any element not recited in that description of the embodiment.
[0058] The term "antibody" herein is used in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity.
[0059] An "antibody fragment" refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds. Examples of antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab')2; diabodies; linear antibodies; single-chain antibody molecules (e.g. scFv); and multispecific antibodies formed from antibody fragments.
[0060] As used herein, the term "prophylactic agent" refers to CD3 binding molecules such as tephzumab which can be used in the prevention, treatment, management or amelioration of one or more symptoms of T1D.
[0061] As used herein, the term "onset" of disease with reference to Type-1 diabetes refers to a patient meeting the criteria established for diagnosis of Type-1 diabetes by the American Diabetes Association (see, Mayfield et al., 2006, Am. Fam. Physician 58: 1355-1362).
[0062] As used herein, the terms "prevent", "preventing" and "prevention" refer to the prevention of the onset of one or more symptoms of T1D in a subject resulting from the administration of a prophy lactic or therapeutic agent.
[0063] As used herein, a "protocol" includes dosing schedules and dosing regimens. The protocols herein are methods of use and include prophylactic and therapeutic protocols. A "dosing regimen" or "course of treatment" may include administration of several doses of a therapeutic or prophylactic agent over 1 to 20 days.
[0064] As used herein, the terms "subject" and "patient" are used interchangeably. As used herein, the terms "subject" and "subjects" refer to an animal, preferably a mammal including a non-primate (e.g., a cow, pig, horse, cat, dog, rat, and mouse) and a primate (e.g., a monkey or a human), and more preferably a human.
[0065] As used herein, the term "prophylactically effective amount" refers to that amount of teplizumab sufficient to result in the delay or prevention of the development, recurrence or onset of one or more symptoms of T1D. In some embodiments, a prophylactically effective amount preferably refers to the amount of teplizumab that delays a subject's onset of T1D by at least 20%, by at least 25%, by at least 30%, by at least 35%, by at least 40%, by at least 45%, by at least 50%, by at least 55%, by at least 60%, by at least 65%, by at least 70%, by at least 75%, by at least 80%, by at least 85%, by at least 90%, by at least 95%.
[0066] Various aspects of the disclosure are described in further detail below. Additional definitions are set out throughout the specification.
Anti-CD3 Antibodies and Pharmaceutical Compositions
[0067] The terms "anti-CD3 antibody" and "an antibody that binds to CD3" refer to an antibody or antibody fragment that is capable of binding cluster of differentiation 3 (CD3) with sufficient affinity such that the antibody is useful as a prophylactic, diagnostic and/or therapeutic agent in targeting CD3. In some embodiments, the extent of binding of an anti-CD3 antibody to an unrelated, non-CD3 protein is less than about 10% of the binding of the antibody to CD3 as measured, e.g., by a radioimmunoassay (RIA). In some embodiments, an antibody that binds to CD3 has a dissociation constant (Kd) of < 1 pM, < 100 nM, < 10 nM, < 1 nM, < 0.1 nM, < 0.01 nM, or < 0.001 nM (e.g. 10’8 M or less, e.g. from 10’8 M to 10’13 M, e.g., from 10’9 M to 10’13 M). In some embodiments, an anti-CD3 antibody binds to an epitope of CD3 that is conserved among CD3 from different species.
[0068] In some embodiments, the anti-CD3 antibody can be ChAglyCD3 (otelixizumab). Otelixizumab is a humanized Fc nonbinding anti-CD3, which was evaluated initially in phase 2 studies by the Belgian Diabetes Registry (BDR) and then developed by Tolerx, which then partnered with GSK to conduct the phase 3 DEFEND new onset T1D trials (NCT00678886, NCT01123083, NCT00763451). Otelixizumab is administered IV with infusions over 8 days. See, e.g., Wiczling et al., J. Clin. Pharmacol. 50 (5) (May 2010) 494-506; Keymeulen et al., N Engl J Med. 2005;352:2598-608; Keymeulen et al., Diabetologia. 2010;53:614-23; Hagopian et al., Diabetes. 2013;62:3901-8; Aronson et al., Diabetes Care. 2014;37:2746-54; Ambery et al., Diabet Med. 2014;31:399-402; Bolt et al., Eur. J. Immunol. 1YY3. 23: 403-411; Vlasakakis et al., Br J Clin Pharmacol (2019) 85 704-714; Guglielmi et al, Expert Opinion on Biological Therapy, 16:6, 841-846; Keymeulen et al., N Engl J Med 2005;352:2598-608; Keymeulen et al., BLOOD 2010, VOL 115, No. 6; Sprangers et al., Immunotherapy (2011) 3(11), 1303-1316; Daifotis et al., Clinical Immunology (2013) 149, 268-278; all incorporated herein by reference.
[0069] In some embodiments, the anti-CD3 antibody can be visilizumab (also called HuM291 ; Nuvion). Visilizumab is a humanized anti-CD3 monoclonal antibody characterized by a mutated IgG2 isotype, lack of binding to Fey receptors, and the ability to induce apoptosis selectively in activated T cells. It was evaluated in patients in graft-versus-host disease (NCT00720629; NCT00032279) and in ulcerative colitis (NCT00267306) and Crohn’s Disease (NCT00267709). See, e.g., Sandborn et al., Gut 59 (11) (Nov 2010) 1485-1492, incorporated herein by reference.
[0070] In some embodiments, the anti-CD3 antibody can be foralumab, a fully human anti- CD3 monoclonal antibody being developed by Tiziana Life Sciences, PLC in NASH and T2D (NCT03291249). See, e.g., Ogura et al., Clin Immunol, 2017;183:240-246; Ishikawa et al., Diabetes. 2007;56(8):2103-9; Wuetal., J Immunol, 2010;185(6):3401-7; all incorporated herein by reference.
[0071] In some embodiments, the anti-CD3 antibody can be teplizumab. Teplizumab, also known as hOKT3yl(Ala-Ala) (containing an alanine at positions 234 and 235) is an anti-CD3 antibody that had been engineered to alter the function of the T lymphocytes that mediate the destruction of the insulin-producing beta cells of the islets of the pancreas. Teplizumab binds to an epitope of the CD3s chain expressed on mature T cells and by doing so changes their function. Sequences and compositions of teplizumab are disclosed in U.S. Patent Nos. 6,491,916; 8,663,634; and 9,056,906, each incorporated herein by reference in its entirety. The full sequences of light and heavy chains are set forth below. Bolded portions are the complementarity determining regions.
Teplizumab Light Chain (SEQ ID NO: 1):
DIQMTQSPSSLSASVGDRVTITCSASSSVSYMNWYQQTPGKAPKRWIYDTSKLASGVP SRFSGSGSGTDYTFTISSLQPEDIATYYCQQWSSNPFTFGQGTKLQITRTVAAPSVFIFP PSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLS
STLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
Teplizumab Heavy Chain (SEQ ID NO: 2):
QVQLVQSGGGVVQPGRSLRLSCKASGYTFTRYTMHWVRQAPGKGLEWIGYINPSRG YTNYNQKVKDRFTISRDNSKNTAFLQMDSLRPEDTGVYFCARYYDDHYCLDYWGQ GTPVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGV HTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHT CPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVE VHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK GQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVL DSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
[0072] In some embodiments, provided herein, is a pharmaceutical composition. Such compositions compnse a prophylactically effective amount of an anti-CD3 antibody, and a pharmaceutically acceptable carrier. In some embodiments, the term "pharmaceutically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term "carrier" refers to a diluent, adjuvant (e.g., Freund's adjuvant (complete and incomplete)), excipient, or vehicle with which the therapeutic is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like (See, for example. Handbook of Pharmaceutical Excipients, Arthur H. Kibbe (ed., 2000, which is incorporated by reference herein in its entirety), Am. Pharmaceutical Association, Washington, D.C.
[0073] The composition, if desired, can also contain minor amounts of weting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained release formulations and the like. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E. W. Martin. Such compositions will contain a prophylactically or therapeutically effective amount of a prophylactic or therapeutic agent preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration. In some embodiments, the pharmaceutical compositions are sterile and in suitable form for administration to a subject, preferably an animal subject, more preferably a mammalian subject, and most preferably a human subject.
[0074] In some embodiments, it may be desirable to administer the pharmaceutical compositions locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion, by injection, or by means of an implant, the implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. Preferably, when administering the anti-CD3 antibody, care must be taken to use materials to which the anti-CD3 antibody does not absorb.
[0075] In some embodiments, the composition can be delivered in a vesicle, in particular a liposome (see Langer, Science 249: 1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353- 365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.).
[0076] In some embodiments, the composition can be delivered in a controlled release or sustained release system. In some embodiments, a pump may be used to achieve controlled or sustained release (see Langer, supra; Sefton, 1987, CRC Crit. Ref. Biomed. Eng. 14:20; Buchwald et al., 1980, Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med. 321 :574). In some embodiments, polymeric materials can be used to achieve controlled or sustained release of the antibodies of the invention or fragments thereof (see e.g., Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds ), Wiley, New York (1984); Ranger and Peppas, 1983, J., Macromol. Sci. Rev. Macromol. Chem. 23:61; see also Levy et al., 1985, Science 228: 190; During et al., 1989, Ann. Neurol. 25:351; Howard et al., 1989, J. Neurosurg. 71: 105); U.S. Pat. No. 5,679,377; U.S. Pat. No. 5,916,597; U.S. Pat. No. 5,912,015; U.S. Pat. No. 5,989,463; U.S. Pat. No. 5,128,326; PCT Publication No. WO 99/15154; and PCT Publication No. WO 99/20253. Examples of polymers used in sustained release formulations include, but are not limited to, poly(2-hydroxy ethyl methacrylate), poly(methyl methacrylate), poly(acrylic acid), poly(ethylene-co-vinyl acetate), poly(methacrylic acid), polyglycolides (PLG), polyanhydrides, poly(N-vinyl pyrrolidone), poly(vinyl alcohol), polyacrylamide, poly(ethylene glycol), polylactides (PLA), poly(lactide-co-glycolides) (PLGA), and polyorthoesters. In some embodiments, the polymer used in a sustained release formulation is inert, free of leachable impurities, stable on storage, sterile, and biodegradable. In some embodiments, a controlled or sustained release system can be placed in proximity of the therapeutic target, i.e., the lungs, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
[0077] Controlled release systems are discussed in the review by Langer (1990, Science 249: 1527-1533). Any technique known to one of skill in the art can be used to produce sustained release formulations comprising one or more antibodies of the invention or fragments thereof. See, e.g., U.S. Pat. No. 4,526,938; PCT Publication No. WO 91/05548; PCT Publication No. WO 96/20698; Ning et al., 1996, Radiotherapy & Oncology 39:179-189; Song et al., 1995, PDA Journal of Pharmaceutical Science & Technology 50:372-397; Cleek et al., 1997, Pro. Infl. Symp. Control. Rel. Bioact. Mater. 24:853-854; and Lam et al., 1997, Proc. Infl. Symp. Control Rel. Bioact. Mater. 24:759-760, each of which is incorporated herein by reference in its entirety.
[0078] A pharmaceutical composition can be formulated to be compatible with its intended route of administration. Examples of routes of administration include, but are not limited to, parenteral, e.g., intravenous, intradermal, subcutaneous, oral, intranasal (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration. In some embodiments, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous, subcutaneous, intramuscular, oral, intranasal or topical administration to human beings. In some embodiments, a pharmaceutical composition is formulated in accordance with routine procedures for subcutaneous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lignocamne to ease pain at the site of the injection.
[0079] The compositions may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. In some embodiments, the disclosure provides dosage forms that permit administration of the anti-CD3 antibody continuously over a period of hours or days (e.g., associated with a pump or other device for such delivery), for example, over a period of 1 hour, 2 hours, 3 hours, 4 hours, 6 hours, 8 hours, 10 hours, 12 hours, 16 hours, 20 hours, 24 hours, 30 hours, 36 hours, 4 days, 5 days, 7 days, 10 days or 14 days. In some embodiments, the invention provides dosage forms that permit administration of a continuously increasing dose, for example, increasing from 51 ug/m2/day to 826 ug/m2/day over a period of 24 hours, 30 hours, 36 hours, 4 days, 5 days, 7 days, 10 days or 14 days.
[0080] The compositions can be formulated as neutral or salt forms. Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, tri ethyl amine, 2-ethylamino ethanol, histidine, procaine, etc.
[0081] Generally, the ingredients of the compositions disclosed herein are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle or bag containing sterile pharmaceutical grade water or saline. In some embodiments, the composition is in solution. In some embodiments, the composition is packaged into a vial (e.g. sterile glass vial of 0.9% sodium chloride injection) or into an infusion bag (e.g. Polyvinylchloride (PVC) infusion bag of 0.9% sodium chloride injection). In some embodiments, the vial is for single-dose. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
[0082] In particular, the disclosure provides that the anti-CD3 antibodies, or pharmaceutical compositions thereof, can be packaged in a hermetically sealed container such as an ampoule or sachette indicating the quantity of the agent. In some embodiments, the anti-CD3 antibody, or pharmaceutical compositions thereof is supplied as a dry sterilized lyophilized powder or water free concentrate in a hermetically sealed container and can be reconstituted, e.g., with water or saline to the appropriate concentration for administration to a subject. Preferably, the anti-CD3 antibody, or pharmaceutical compositions thereof is supplied as a dry sterile lyophilized powder in a hermetically sealed container at a unit dosage of at least 5 mg, more preferably at least 10 mg, at least 15 mg, at least 25 mg, at least 35 mg, at least 45 mg, at least 50 mg, at least 75 mg, or at least 100 mg. The lyophilized prophylactic agents, or pharmaceutical compositions herein should be stored at between 2 °C and 8 °C in its original container and the prophylactic or therapeutic agents, or pharmaceutical compositions of the invention should be administered within 1 week, preferably within 5 days, within 72 hours, within 48 hours, within 24 hours, within 12 hours, within 6 hours, within 5 hours, within 3 hours, or within 1 hour after being reconstituted. In some embodiments, the pharmaceutical composition is supplied in liquid form in a hermetically sealed container indicating the quantity and concentration of the agent. Preferably, the liquid fonn of the administered composition is supplied in a hermetically sealed container at least 0.25 mg/ml, more preferably at least 0.5 mg/ml, at least 1 mg/ml, at least 2.5 mg/ml, at least 5 mg/ml, at least 8 mg/ml, at least 10 mg/ml, at least 15 mg/ml, at least 25 mg/ml, at least 50 mg/ml, at least 75 mg/ml or at least 100 mg/ml. The liquid form should be stored at between 2 °C and 8 °C in its original container.
[0083] In some embodiments, the disclosure provides that the composition of the invention is packaged in a hennetically sealed container such as an ampoule or sachette indicating the quantity of the anti-CD3 antibody.
[0084] The compositions may, if desired, be presented in a pack or dispenser device that may contain one or more unit dosage forms containing the active ingredient. The pack may, for example, comprise metal or plastic foil, such as a blister pack.
[0085] The amount of the composition of the invention which will be effective in the prevention or amelioration of one or more symptoms associated with T1D can be determined by standard clinical techniques. The precise dose to be employed in the formulation will also depend on the route of administration and the seriousness of the condition, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
Methods and Use
[0086] In some embodiments, the present disclosure encompasses administration of antihuman CD3 antibodies such as teplizumab to individuals predisposed to develop type 1 diabetes or with pre-clinical stages of type 1 diabetes, but who do not meet the diagnosis criteria as established by the American Diabetes Association or the Immunology of Diabetes Society to prevent or delay the onset of type 1 diabetes and/or to prevent or delay the need for administration of insuhn to such patients. In some embodiments, high-risk factors for identification of predisposed subjects include having first or second degree relatives with diagnosed type-1 diabetes, an impaired fasting glucose level (e.g., at least one determination of a glucose level of 100-125 mg/dl after fasting (8 hours with no food)), an impaired glucose tolerance in response to a 75 g OGTT (e.g., at least one determination of a 2-hr glucose level of 140-199 mg/dl in response to a 75 g OGTT), an HLA type of DR3, DR4 or DR7 in a Caucasian, an HLA type of DR3 or DR4 in a person of African descent, an HLA type of DR3, DR4 or DR9 in a person of Japanese descent, exposure to viruses (e.g., coxsackie B virus, enteroviruses, adenoviruses, rubella, cytomegalovirus, Epstein-Barr vims), a positive diagnosis according to art accepted criteria of at least one other autoimmune disorder (e.g., thyroid disease, celiac disease), and/or the detection of autoantibodies, particularly ICAs and type 1 diabetes-associated autoantibodies, in the serum or other tissues. In some embodiments, the subject identified as predisposed to developing type 1 diabetes has at least one of the risk factors described herein and/or as known in the art. The present disclosure also encompasses identification of subjects predisposed to development of type 1 diabetes, wherein the subject presents a combination of two or more, three or more, four or more, or more than five of the risk factors disclosed herein or known in the art. In some embodiments, the non-diabetic subject is an adult or a pediatric subject. In some embodiments, the pediatric subject is 8 years old or older. In some embodiments, the pediatric subject is 7 years old or older. In some embodiments, the pediatric subject is 6 years old or older. In some embodiments, the pediatric subject is 5 years old or older. In some embodiments, the pediatric subject is 4 years old or older. In some embodiments, the pediatric subject is 3 years old or older. In some embodiments, the pediatric subject is 2 years old or older. In some embodiments, the pediatric subject is 1 year old or older. In some embodiments, the pediatric subject is an infant. In some embodiments, the infant is 12 months old or less, 11 months old or less, 10 months old of less, 9 months old or less, 8 months old or less, 7 months old or less, 6 months old or less, 5 months old or less, 4 months old or less, 3 months old or less, 2 months old or less, I month old or less.
[0087] Serum autoantibodies associated with type 1 diabetes or with a predisposition for the development of type 1 diabetes are islet-cell autoantibodies (e.g., anti-ICA512 autoantibodies), glutamic acid decarbamylase autoantibodies (e.g., anti-GAD65 autoantibodies), IA2 antibodies, ZnT8 antibodies and/or anti-insulin autoantibodies. Accordingly, in a specific example in accordance with this embodiment, the invention encompasses the treatment of an individual with detectable autoantibodies associated with a predisposition to the development of type 1 diabetes or associated with early stage type 1 diabetes (e.g., anti-IA2, anti-ICA512, anti-GAD or antiinsulin autoantibodies), wherein the individual has not been diagnosed with type 1 diabetes and/or is a first or second degree relative of a type-1 diabetic. In some embodiments, the presence of the autoantibodies is detected by ELISA, electrochemoluminescence (ECL), radioassay (see, e.g., Yu et al., 1996, J. Clin. Endocrinol. Metab. 81:4264-4267), agglutination PCR (Tsai et al, ACS Central Science 2016 2 (3), 139-147) or by any other method for immunospecific detection of antibodies described herein or as known to one of ordinary skill in the art.
[0088] P-cell function prior to, during, and after therapy may be assessed by methods described herein or by any method known to one of ordinary skill in the art. For example, the Diabetes Control and Complications Trial (DCCT) research group has established the monitoring of percentage glycosylated hemoglobin (HA1 and HAlc) as the standard for evaluation of blood glucose control (DCCT, 1993, N. Engl. J. Med. 329:977-986). Alternatively, characterization of daily insulin needs, C-peptide levels/response, hypoglycemic episodes, and/or FPIR may be used as markers of p-cel 1 function or to establish a therapeutic index (See Keymeulen et al, 2005, N. Engl. J. Med. 352:2598-2608; Herold et al, 2005, Diabetes 54: 1763- 1769; U.S. Pat. Appl. Pub. No. 2004/0038867 Al; and Greenbaum et al, 2001, Diabetes 50:470- 476, respectively). For example, FPIR is calculated as the sum of insulin values at 1 and 3 minutes post IGTT, which are performed according to Islet Cell Antibody Register User's Study protocols (see, e.g., Bingley et al, 1996, Diabetes 45: 1720-1728 and McCulloch et al., 1993, Diabetes Care 16:911-915).
[0089] In some embodiments, the individuals predisposed to develop T1D can be a nondiabetic subject who is a relative of a patient with T1D. In some embodiments, the non-diabetic subject has 2 or more diabetes-related autoantibodies selected from islet cell antibodies (ICA), insulin autoantibodies (IAA), and antibodies to glutamic acid decarboxylase (GAD), tyrosine phosphatase (IA-2/ICA512) or ZnT8.
[0090] In some embodiments, the non-diabetic subj ect has abnormal glucose tolerance on oral glucose tolerance test (OGTT). Abnormal glucose tolerance on OGTT is defined as a fasting glucose level of 110-125 mg/dL, or 2 hour plasma of > 140 and < 200 mg/dL, or an intervening glucose value at 30, 60, or 90 minutes on OGTT > 200 mg/dL. [0091] In some embodiments, the non-diabetic subject has abnormal results in a glucose monitoring system (CGM) revealing high sensor average glucose levels (>=110 mg/dL), or high variability of glycemia (CV >=15), or less time in range (>=10% of the time above 140 mg/dL).
[0092] In some embodiments, the non-diabetic subject who will respond to the anti-CD3 antibody such as teplizumab does not have antibodies against ZnT8. In some embodiments, such non-diabetic subject is HLA-DR4+ and is not HLA-DR3+. In some embodiments, such non- diabetic subject who will respond to the anti-CD3 antibody such as teplizumab demonstrates an increase, following administration (e.g., after 1 month, after 2 months, after 3 months, or longer or shorter), in the frequency (or relative amount) of TIGIT+KLRG1+CD8+ T-cells (e.g., by flow cytometry) in peripheral blood mononuclear cells.
[0093] In some embodiments, the methods provided herein comprise administering an antihuman CD3 antibodies such as teplizumab to a subject in need thereof with Stage 2 type 1 diabetes. In some embodiments, the subject is an adult. In some embodiments, the subject is a pediatric subject. In some embodiments, the pediatric subject is 8 years old or older (e.g. 8 years old, 9 years old, 10 years old, 11 years old, 12 years old, 13 years old, 14 years old, 15 years old, 16 years old, 17 years old, 18 years old or older). In some embodiments, administration of the anti-human CD3 antibodies such as teplizumab delay the onset of Stage 3 Type 1 diabetes. In some embodiments, the subject has Stage 2 type 1 diabetes. In some embodiments, the subject that has dysglycemia without overt hyperglycemia has at least two positive pancreatic islet cell autoantibodies. See ADA and ISPAD guidelines which define classification and diagnosis of Stage 2 T1D in American Diabetes Association Professional Practice Committee- Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes-2022. Diabetes Care. 2022 Jan l;45(Suppl 1):S 17-S38); and in Besser REJ, Bell KJ, Couper JJ, Ziegler AG, Wherrett DK, Knip M, Speake C, Casteels K, Driscoll KA, Jacobsen L, Craig ME, Haller MJ. ISPAD clinical practice consensus guidelines 2022: Stages of type 1 diabetes in children and adolescents. Pediatr Diabetes. 2022 Sep 30). In some embodiments, the subject has abnormal glucose tolerance on oral glucose tolerance test (OGTT). In some embodiments, the subject in need thereof meets criteria for a diagnosis of Stage 2 type 1 diabetes does not have type 2 diabetes. In some embodiments, the methods provided herein comprise confirming that the subject who has dysglycemia without overt hyperglycemia has Stage 2 type 1 diabetes by documenting at least two positive pancreatic islet cell autoantibodies. In some embodiments, the methods provided herein further comprise confirming that the subject does not have type 2 diabetes. In some embodiments, subjects having Stage 2 type 1 diabetes have both of the following:
1. Two or more of the following pancreatic islet autoantibodies:
Glutamic acid decarboxylase 65 (GAD) autoantibodies
Insulin autoantibody (IAA)
Insulinoma-associated antigen 2 autoantibody (IA-2A)
Zinc transporter 8 autoantibody (ZnT8A)
Islet cell autoantibody (ICA)
2. Dysglycemia (e g. on oral glucose tolerance testing).
[0094] In some embodiments, Stage 2 type 1 diabetes can be diagnosed in other ways known to those skilled in the art.
[0095] Aspects of the disclosure relate to methods of preventing or delaying onset of Stage 3 type 1 diabetes (T1D). In some embodiments, the subject is an adult. In some embodiments, the subject is a child 1 year old, 2 years old, 3 years old or older, 4 years old and older, 5 years old and older, 6 years old and older, 7 years old and older, 8 years old and older, 9 years old and older, 10 years old and older, 11 years old and older, 12 years old and older, 13 years old and older, 14 years old and older, 15 years old and older, 16 years old and older, 17 years old and older, 18 years old and older. In some embodiments, the subject is a infant ( 12 months old or less, 11 months old or less, 10 months old or less, 9 months old or less, 8 months old or less, 7 months old or less, 6 months old or less, 5 months old or less, 4 months old or less, 3 months old or less, 2 months old or less, 1 month old or less (e.g. initial weeks of life)).
[0096] In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at a cumulative dose greater than 10,000 micrograms/meter squared (pg/m2). In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 9,500 to about 14,000 pg/m2, from about 9,500 to about 13,500 pg/m2, from about 9,500 to about 13,000 pg/ m2, from about 9,500 to about 12,500 pg/m2, from about 9,500 to about 12,000 pg/m2, from about 9,500 to about 11,500 pg/ m2, from about 9,500 to about 11,000 pg/m2, from about 9,500 to about 10,500 pg/m2, from about 9,500 to about 10,000 pg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 10,000 to about 14,000 pg/m2, from about 10,000 to about 13,500 pg/ m2, from about 10,000 to about 13,000 pg/m2, from about 10,000 to about 12,500 pg/m2, from about 10,000 to about 12,000 pg/m2, from about 10,000 to about 11,500 pg/m2, from about 10,000 to about 11,000 pg/ m2, from about 10,000 to about 10,500 pg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 10,500 to about 14,000 pg/m2, from about 10,500 to about 13,500 pg/m2, from about 10,500 to about 13,000 pg/m2, from about 10,500 to about 12,500 pg/m2, from about 10,500 to about 12,000 pg/m2, from about 10,500 to about 11,500 pg/ m2, from about 10,500 to about 11,000 pg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 11,000 to about 14,000 pg/m2, from about 11,000 to about 13,500 pg/m2, from about 11,000 to about 13,000 pg/m2, from about 11,000 to about 12,500 pg/m2, from about 11,000 to about 12,000 pg/m2, from about 11,000 to about 11,500 pg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 11,000 to about 14,000 pg/m2, from about 11,000 to about 13,500 pg/ m2, from about 11,000 to about 13,000 pg/m2, from about 11,000 to about 12,500 pg/m2, from about 11,000 to about 12,000 pg/m2, from about 11,000 to about 11,500 pg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 11,500 to about 14,000 pg/m2, from about 11,500 to about 13,500 pg/m2, from about 11,500 to about 13,000 pg/m2, from about 11,500 to about 12,500 pg/m2, from about 11,500 to about 12,000 pg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 12,000 to about 14,000 pg/m2, from about 12,000 to about 13,500 pg/m2, from about 12,000 to about 13,000 pg/m2, from about 12,000 to about 12,500 pg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 12,500 to about 14,000 pg/m2, from about 12,500 to about 13,500 pg/m2, from about 12,500 to about 13,000 pg/m2. In some embodiments, the prophy tactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 13,000 to about 14,000 pg/m2, from about 13,000 to about 13,500 pg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at from about 13,500 pg/m2 to about 14,000 pg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14 days course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody at about 10,000 pg/m2, 10,500 pg/m2, 11,000 pg/m2, 11.500 pg/m2, 12,000 pg/m2, 12,500 pg/m2, 13,000 pg/m2, 13,500 pg/m2, or 14,000 pg/m2. In some embodiments, the anti-CD3 antibody is or comprises teplizumab.
[0097] In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14 days course of intravenous (IV) infusion of teplizumab at from about 10,000 to about 14,000 pg/m2, from about 10,000 to about 13,500 pg/m2, from about 10,000 to about 13,000 pg/m2, from about 10,000 to about 12,500 pg/m2, from about 10,000 to about 12,000 pg/m2, from about 10,000 to about 11,500 pg/ m2, from about 10,000 to about 11,000 pg/m2, from about 10,000 to about 10,500 pg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14 days course of intravenous (IV) infusion of teplizumab at from about 10,500 to about 14,000 pg/m2, from about 10,500 to about 13,500 pg/m2, from about 10,500 to about 13,000 pg/m2, from about 10,500 to about 12,500 pg/m2, from about 10,500 to about 12,000 pg/m2, from about 10,500 to about 11,500 pg/m2, from about 10,500 to about 11,000 pg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises a 10 to 14 days course of intravenous (IV) infusion of teplizumab at from about 11 ,000 to about 14,000 pg/m2, from about 11,000 to about 13,500 pg/m2, from about 11,000 to about 13,000 pg/m2, from about 11,000 to about 12,500 pg/m2, from about 11,000 to about 12,000 pg/m2, from about 11,000 to about 11,500 pg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises 10 to 14 days course of intravenous (IV) infusion of teplizumab at from about 11,000 to about 14,000 pg/m2, from about 11,000 to about 13,500 pg/m2, from about 11,000 to about 13,000 pg/m2, from about 11,000 to about 12,500 pg/m2, from about 11,000 to about 12,000 pg/m2, from about 11,000 to about 11,500 pg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises 10 to 14 days course of intravenous (IV) infusion of teplizumab at from about 11,500 to about 14,000 pg/m2, from about 11,500 to about 13,500 gg/m2, from about 11,500 to about 13,000 gg/m2, from about
11.500 to about 12,500 gg/m2, from about 11,500 to about 12,000 gg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises 10 to 14 days course of intravenous (IV) infusion of tephzumab at from about 12,000 to about 14,000 gg/m2, from about 12,000 to about 13,500 gg/m2, from about 12,000 to about 13,000 gg/m2, from about 12,000 to about 12,500 gg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises 10 to 14 days course of intravenous (IV) infusion of teplizumab at from about 12,500 to about 14,000 gg/m2, from about 12,500 to about 13,500 gg/m2, from about
12.500 to about 13,000 gg/ m2. In some embodiments, the prophylactically effective amount of the antibody comprises 10 to 14 days course of intravenous (IV) infusion of teplizumab at from about 13,000 to about 14,000 gg/m2, from about 13,000 to about 13,500 gg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises 10 to 14 days course of intravenous (IV) infusion of teplizumab at from about 13,500 gg/m2 to about 14,000 gg/m2. In some embodiments, the prophylactically effective amount of the antibody comprises 10 to 14 days course of intravenous (IV) infusion of teplizumab at about 10,000 gg/ m2, 10,500 gg/m2, 11,000 gg/m2, 11.500 gg/m2, 12,000 gg/m2, 12,500 gg/m2, 13,000 gg/m2, 13,500 gg/m2, or 14,000 gg/m2. In some embodiments, the anti-CD3 antibody is or comprises teplizumab.
[0098] In some embodiments, the method comprises administering to the non-diabetic subj ect who is at risk for T1D a 14-day course IV infusion at about 60 gg/m2, about 125 gg/m2, about 250 gg/m2, and about 500 gg/m2, on days 1-4, respectively, and a dose of about 1,000 gg/m2 on each of days 5-14. In some embodiments, the cumulative dose is about 10,935 gg/m2.
[0099] In some embodiments, the method comprises administering to the non-diabetic subj ect who is at risk for T1D a 14-day course IV infusion at about 60 gg/m2, about 125 gg/m2, about 250 gg/m2, and about 500 gg/m2, on days 1-4, respectively, and a dose of about 1,030 gg/m2 on each of days 5-14. In some embodiments, the cumulative dose is about 11,235 gg/m2.
[00100] In some embodiments, the method comprises administering to the non-diabetic subject who is at risk for T1D a 14-day course IV infusion at about 100 gg/m2, about 425 gg/m2, about 850 gg/m2, and about 850 gg/m2, on days 1-4, respectively, and a dose of about 1,000 gg/m2 on each of days 5-14. In some embodiments, the cumulative dose is about 12,225 gg/m2.
[00101] In some embodiments, the method comprises administering to the non-diabetic subject who is at risk for T1D a 14-day course IV infusion at about 65 gg/m2, about 125 gg/m2, about 250 pg/m2, and about 500 pg/m2, on days 1-4, respectively, and a dose of about 1,070 pg/m2 on each of days 5-14. In some embodiments, the cumulative dose is about 11,640 pg/m2.
[00102] In some embodiments, the method comprises administering to a subject in need thereof a 14-day course IV infusion, wherein the daily dose is at about 65 pg/m2, about 125 pg/m2, about 250 pg/m2, and about 500 pg/m2, on days 1-4, respectively, and at about 1,030 pg/m2 on each of days 5-14. In some embodiments, the cumulative dose is about 11,240 pg/m2.
[00103] In some embodiments, the method comprises administering to a subject in need thereof a 14-day course IV infusion, wherein the daily dose is at about 60 pg/m2, about 125 pg/m2, about 250 pg/m2, and about 500 pg/m2, on days 1-4, respectively, and at about 1,000 pg/m2 on each of days 5-14. In some embodiments, the cumulative dose is about 10,935 pg/m2.
[00104] In some embodiments, the method comprises administering to a subject in need thereof a 14-day course IV infusion, wherein the daily dose is at about 60 pg/m2, about 125 pg/m2, about 250 pg/m2, and about 500 pg/m2, on days 1-4, respectively, and at about 1,030 pg/m2 on each of days 5-14. In some embodiments, the cumulative dose is about 11,235 pg/m2.
[00105] In some embodiments, the method comprises administering to subject in need therof a 14-day course IV infusion, wherein the daily dose is at about 100 pg/m2, about 425 pg/m2, about 850 pg/m2, and about 850 pg/m2, on days 1-4, respectively, and at about 1,000 pg/m2 on each of days 5-14. In some embodiments, the cumulative dose is about 12,225 pg/m2.
[00106] In some embodiments, the method comprises administering to a subject in need thereof a 14-day course IV infusion, wherein the daily dose is at about 65 pg/m2, about 125 pg/m2, about 250 pg/m2, and about 500 pg/m2, on days 1-4, respectively, and a dose of about 1,070 pg/m2 on each of days 5-14. In some embodiments, the cumulative dose is about 11,640 pg/m2.
[00107] In some embodiments, the method comprises administering to a subject in need thereof a 14-day course IV infusion, wherein the daily dose is at about 65 pg/m2, about 125 pg/m2, about 250 pg/m2, and about 500 pg/m2, on days 1-4, respectively, and at about 1,030 pg/m2 on each of days 5-14. In some embodiments, the cumulative dose is about 11,240 pg/m2.
[00108] In some embodiment, the anti-CD3 antibody daily administration is missed, and dosing resumes by administering all remaining doses on consecutive days to complete the 14-day treatment course.
[00109] In some embodiments, the prophylactically effective amount delays median time to clinical diagnosis of T1D by at least 50%, at least 80%, or at least 90%, or at least 12 months, at least 18 months, at least 24 months, at least 36 months, at least 48 months, or at least 60 months, or longer.
[00110] In some embodiments, the course of dosing with the anti-CD3 antibody such as teplizumab can be repeated at 2 month, 4 month, 6 month, 8 month, 9 month, 10 month, 12 month, 15 month, 18 month, 24 month, 30 month, or 36 month intervals. In some embodiments, efficacy of the treatment with the anti-CD3 antibody such as teplizumab is determined as described herein, or as is known in the art, at 2 months, 4 months, 6 months, 9 months, 12 months, 15 months, 18 months, 24 months, 30 months, or 36 months subsequent to the previous treatment.
[00111] In some embodiments, a subject is administered one or more unit doses of approximately 0.5-50 pg/kg, approximately 0.5-40 pg/kg, approximately 0.5-30 pg/kg, approximately 0.5-20 pg/kg, approximately 0.5-15 pg/kg, approximately 0.5-10 pg/kg, approximately 0.5-5 pg/kg, approximately 1-5 pg/kg, approximately 1-10 pg/kg, approximately 20-40 pg/kg, approximately 20-30 pg/kg, approximately 22-28 pg/kg or approximately 25-26 pg/kg of the anti-CD3 antibody such as teplizumab to prevent, treat, ameliorate, mitigate, relieve, lighten, postpone, stop or halt one or more symptoms of T1D. In some embodiments, a subject is administered one or more unit doses of about 200 pg/kg, 178 pg/kg, 180 pg/kg, 128 pg/kg, 100 pg/kg, 95 pg/kg, 90 pg/kg, 85 pg/kg, 80 pg/kg, 75 pg/kg, 70 pg/kg, 65 pg/kg, 60 pg/kg, 55 pg/kg, 50 pg/kg, 45 pg/kg, 40 pg/kg, 35 pg/kg, 30 pg/kg, 26 pg/kg, 25 pg/kg, 20 pg/kg, 15 pg/kg, 13 pg/kg, 10 pg/kg, 6.5 pg/kg, 5 pg/kg, 3.2 pg/kg, 3 pg/kg, 2.5 pg/kg, 2 pg/kg, 1.6 pg/kg, 1.5 pg/kg, 1 pg/kg, 0.5 pg/kg, 0.25 pg/kg, 0.1 pg/kg, or 0.05 pg/kg of the anti-CD3 antibody such as teplizumab to prevent, treat, ameliorate, mitigate, relieve, lighten, postpone, stop or halt one or more symptoms of T1D.
[00112] In some embodiments, a subject is administered one or more doses of the anti-CD3 antibody such as teplizumab at about 5-1200 pg/m2, for example, at about 60-1070 pg/m2. In some embodiments, a subject is administered one or more unit doses of 1200 pg/m2, 1150 pg/m2, 1100 pg/m2, 1050 pg/m2, 1000 pg/m2, 950 pg/m2, 900 pg/m2, 850 pg/m2, 800 pg/m2, 750 pg/m2, 700 pg/m2, 650 pg/m2, 600 ug/m2, 550 pg/m2, 500 pg/m2, 450 pg/m2, 400 pg/m2, 350 pg/m2, 300 pg/m2, 250 pg/m2, 200 pg/m2, 150 pg/m2, 100 pg/m2, 50 pg/m2, 40 pg/m2, 30 pg/m2, 20 pg/m2, 15 pg/m2, 10 pg/m2, or 5 pg/m2 of the anti-CD3 antibody such as teplizumab to prevent, treat, slow the progression of, delay the onset of T1D, ameliorate, mitigate, relieve, lighten, postpone, stop or halt one or more symptoms of T1D. [00113] In some embodiments, the subject is administered a treatment regimen comprising one or more doses of a prophylactically effective amount of the anti-CD3 antibody such as teplizumab, wherein the course of treatment is administered over 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days or 14 days. In some embodiments, the subject is administered a treatment regimen comprising one or more doses of a prophylactically effective amount of the anti-CD3 antibody such as teplizumab, wherein the course of treatment is administered over 14 days. In some embodiments, the treatment regimen comprises administering doses of the prophylactically effective amount every day, every 2nd day, every 3rd day or every 4th day. In some embodiments, the treatment regimen comprises administering doses of the prophylactically effective amount on Monday, Tuesday, Wednesday, Thursday of a given week and not administering doses of the prophylactically effective amount on Friday, Saturday, and Sunday of the same week until 14 doses, 13 doses, 12 doses, 11 doses, 10 doses, 9 doses, or 8 doses have been administered. In some embodiments, the dose administered is the same each day of the regimen.
[00114] In some embodiments, a subject is administered a treatment regimen comprising one or more doses of a prophylactically effective amount of the anti-CD3 antibody such as teplizumab, wherein the prophylactically effective amount is about 200 pg/kg /day, 175 pg/kg/day, 150 pg/kg/day, 125 pg/kg/day, 100 pg/kg/day, 95 pg/kg/day, 90 pg/kg/day, 85 pg/kg/day, 80 pg/kg/day, 75 pg/kg/day, 70 pg/kg/day, 65 pg/kg/day, 60 pg/kg/day, 55 pg/kg/day, 50 pg/kg/day, 45 pg/kg/day, 40 pg/kg/day, 35 pg/kg/day, 30 pg/kg/day, 26 pg/kg/day, 25 pg/kg/day, 20 pg/kg/day, 15 pg/kg/day, 13 pg/kg/day, 10 pg/kg/day, 6.5 pg/kg/day, 5 pg/kg/day, 3.2 pg/kg/day, 3 pg/kg/day, 2.5 pg/kg/day, 2 pg/kg/day, 1.6 pg/kg/day, 1.5 pg/kg/day, 1 pg/kg/day, 0.5 pg/kg/day, 0.25 pg/kg/day, 0.1 pg/kg/day, or 0.05 pg/kg/day; and/or wherein the prophylactically effective amount is about 1200 pg/m2/day, 1150 pg/m2/day, 1100 pg/m2/day, 1050 pg/m2/day, 1000 pg/m2/day, 950 pg/m2/day, 900 pg/m2/day, 850 pg/m2/day, 800 pg/m2/day, 750 pg/m2/day, 700 pg/m2/day, 650 pg/m2/day, 600 pg/m2/day, 550 pg/m2/day, 500 pg/m2/day, 450 pg/m2/day, 400 pg/m2/day, 350 pg/m2/day, 300 pg/m2/day, 250 pg/m2/day, 200 pg/m2/day, 150 pg/m2/day, 100 pg/m2/day, 50 pg/m2/day, 40 pg/m2/day, 30 pg/m2/day, 20 pg/m2/day, 15 pg/m2/day, 10 pg/m2/day, or 5 pg/m2/day.
[00115] In some embodiments, the intravenous dose of about 1200 pg/m2 or less, 1150 pg/m2 or less, 1100 pg/m2 or less, 1050 pg/m2 or less, 1000 pg/m2 or less, 950 pg/m2 or less, 900 pg/m2 or less, 850 pg/m2 or less, 800 pg/m2 or less, 750 pg/m2 or less, 700 pg/m2 or less, 650 pg/m2 or less, 600 gg/m2 or less, 550 gg/m2 or less, 500 gg/m2or less, 450 gg/m2 or less, 400 gg/m2 or less, 350 gg/m2 or less, 300 gg/m2 or less, 250 gg/m2 or less, 200 gg/m2 or less, 150 gg/m2 or less, 100 gg/m2 or less, 50 gg/m2 or less, 40 gg/m2 or less, 30 gg/m2 or less, 20 gg/m2 or less, 15 gg/m2 or less, 10 gg/m2 or less, or 5 gg/m2 or less of the anti-CD3 antibody such as teplizumab is administered over about 24 hours, about 22 hours, about 20 hours, about 18 hours, about 16 hours, about 14 hours, about 12 hours, about 10 hours, about 8 hours, about 6 hours, about 4 hours, about 2 hours, about 1.5 hours, about 1 hour, about 50 minutes, about 40 minutes, about 30 minutes, about 20 minutes, about 10 minutes, about 5 minutes, about 2 minutes, about 1 minute, about 30 seconds or about 10 seconds to prevent, treat, ameliorate, mitigate, relieve, lighten, postpone, stop or halt one or more symptoms of type 1 diabetes. The total dosage over the duration of the regimen can be greater than about 9,500, 10,000, 10,500, 11,000, 11,500, 12,000, 12,500, 13,000 or 13,500 ug/m2. The total dosage over the duration of the regimen, in some embodiments, is from about 9,500 to about 14,000 gg/m2, from about 9,500 to about
13.500 gg/m2, from about 9,500 to about 13,000 gg/m2, from about 9,500 to about 12,500 gg/m2, from about 9,500 to about 12,000 gg/m2, from about 9,500 to about 11,500 gg/m2, from about
9.500 to about 11,000 gg/m2, from about 9,500 to about 10,500 gg/m2, from about 9,500 to about 10,000 gg/m2. The total dosage over the duration of the regimen, in some embodiments, is from about 10,500 to about 14,000 gg/m2, from about 10,500 to about 13,500 gg/m2, from about
10.500 to about 13,000 gg/m2, from about 10,500 to about 12,500 gg/m2, from about 10,500 to about 12,000 gg/m2, from about 10,500 to about 11,500 gg/m2, from about 10,500 to about 11,000 gg/m2. For example, the total dosage over the duration of the regimen is about 10,935 gg/m2, about 11,235 gg/m2, about 11,240 gg/m2, about 11,640 gg/m2, or about 12,225 gg/m2
[00116] In some embodiments, the dose escalates over the first fourth, first half or first 2/3 of the doses (e.g., over the first 2, 3, 4, 5, or 6 days of a 10, 12, 14, 16, 18 or 20-day regimen of one dose per day) of the treatment regimen until the daily prophylactically effective amount of the anti-CD3 antibody such as teplizumab is achieved. For example, the dose can escalate over 3 or 4 days of a 14-day regimen. In some embodiments, a subject is administered a treatment regimen comprising one or more doses of a prophylactically effective amount of the anti-CD3 antibody such as teplizumab, wherein the prophylactically effective amount is increased by, e g., about 0.01 gg/kg, 0.02 gg/kg, 0.04 gg /kg, 0.05 gg/kg, 0.06 gg/kg, 0.08 gg/kg, 0.1 gg/kg, 0.2 gg/kg, 0.25 gg /kg, 0.5 gg /kg, 0.75 gg /kg, 1 gg /kg, 1.5 gg /kg, 2 gg /kg, 4 gg/kg, 5 gg/kg, 10 gg/kg, 15 gg /kg, 20 gg /kg, 25 gg /kg, 30 gg /kg, 35 gg /kg, 40 gg /kg, 45 gg /kg, 50 gg /kg, 55 gg /kg, 60 gg /kg, 65 gg /kg, 70 gg /kg, 75 gg /kg, 80 gg /kg, 85 gg /kg, 90 gg /kg, 95 gg /kg, 100 gg /kg, or 125 pg /kg each day; or increased by, e.g., 1 pg/m2, 5 pg/m2, 10 gg/m2, 15 gg/m2, 20 gg/m2, 30 gg/m2, 40 gg/m2, 50 gg/m2, 60 gg/m2, 70 gg/m2, 80 gg/m2, 90 gg/m2, 100 gg/m2, 150 gg/m2, 200 gg/m2, 250 gg/m2, 300 gg/m2, 350 gg/m2, 400 gg/m2, 450 gg/m2, 500 gg/m2, 550 gg/m2, 600 gg/m2, or 650 gg/m2, each day as treatment progresses. In some embodiments, a subject is administered a treatment regimen comprising one or more doses of a prophylactically effective amount of the anti-CD3 antibody such as teplizumab, wherein the prophylactically effective amount is increased by a factor of 1.25, a factor of 1.5, a factor of 2, a factor of 2.25, a factor of 2.5, or a factor of 5 until the daily prophylactically effective amount of the anti-CD3 antibody such as teplizumab is achieved.
[00117] In some embodiments, a subject is intramuscularly administered one or more doses of a about 200 gg/kg or less, preferably about 175 gg/kg or less, 150 gg/kg or less, 125 gg/kg or less, 100 gg/kg or less, 95 gg/kg or less, 90 gg/kg or less, 85 gg/kg or less, 80 gg/kg or less, 75 gg/kg or less, 70 gg/kg or less, 65 gg/kg or less, 60 gg/kg or less, 55 gg/kg or less, 50 gg/kg or less, 45 gg/kg or less, 40 gg/kg or less, 35 gg/kg or less, 30 gg/kg or less, 25 gg/kg or less, 20 gg/kg or less, 15 gg/kg or less, 10 gg/kg or less, 5 gg/kg or less, 2.5 gg/kg or less, 2 gg/kg or less, 1.5 gg/kg or less, 1 gg/kg or less, 0.5 gg/kg or less, or 0.2 gg/kg or less of the anti-CD3 antibody such as teplizumab, otelixizumab or foralumab, to prevent, treat, ameliorate, mitigate, relieve, lighten, postpone, stop or halt one or more symptoms of T1D.
[00118] In some embodiments, a subject is subcutaneously administered one or more doses of a about 200 ug/kg or less, preferably about 175 gg/kg or less, 150 gg/kg or less, 125 gg/kg or less, 100 gg/kg or less, 95 gg/kg or less, 90 gg/kg or less, 85 gg/kg or less, 80 gg/kg or less, 75 gg/kg or less, 70 gg/kg or less, 65 gg/kg or less, 60 gg/kg or less, 55 gg/kg or less, 50 gg/kg or less, 45 gg/kg or less, 40 gg/kg or less, 35 gg/kg or less, 30 gg/kg or less, 25 gg/kg or less, 20 gg/kg or less, 15 gg/kg or less, 10 gg/kg or less, 5 gg/kg or less, 2.5 gg/kg or less, 2 gg/kg or less, 1.5 gg/kg or less, 1 gg/kg or less, 0.5 gg/kg or less, or 0.2 gg/kg or less of the anti-CD3 antibody such as teplizumab, otelixizumab or foralumab, to prevent, treat, ameliorate, mitigate, relieve, lighten, postpone, stop or halt one or more symptoms of T1D.
[00119] In some embodiments, a subject is intravenously administered one or more doses of a about 100 ug/kg or less, preferably about 95 ug/kg or less, 90 ug/kg or less, 85 ug/kg or less, 80 ug/kg or less, 75 ug/kg or less, 70 ug/kg or less, 65 ug/kg or less, 60 ug/kg or less, 55 ug/kg or less, 50 ug/kg or less, 45 ug/kg or less, 40 ug/kg or less, 35 ug/kg or less, 30 ug/kg or less, 25 ug/kg or less, 20 ug/kg or less, 15 ug/kg or less, 10 ug/kg or less, 5 ug/kg or less, 2.5 ug/kg or less, 2 ug/kg or less, 1.5 ug/kg or less, 1 ug/kg or less, 0.5 ug/kg or less, or 0.2 ug/kg or less of the anti-CD3 antibody such as teplizumab, otelixizumab or foralumab, to prevent, treat, ameliorate, mitigate, relieve, lighten, postpone, stop or halt one or more symptoms of T1D. In some embodiments, the intravenous dose of about 100 pg/kg or less, 95 pg/kg or less, 90 pg/kg or less, 85 pg/kg or less, 80 pg/kg or less, 75 pg/kg or less, 70 pg/kg or less, 65 u pg/kg or less, 60 pg/kg or less, 55 pg/kg or less, 50 pg/kg or less, 45 pg/kg or less, 40 pg/kg or less, 35 pg/kg or less, 30 pg/kg or less, 25 pg/kg or less, 20 pg/kg or less, 15 pg/kg or less, 10 pg/kg or less, 5 pg/kg or less, 2.5 pg/kg or less, 2 pg/kg or less, 1.5 pg/kg or less, 1 pg/kg or less, 0.5 pg/kg or less, or 0.2 pg/kg or less of the anti-CD3 antibody such as teplizumab, otelixizumab or foralumab, is administered over about 6 hours, about 4 hours, about 2 hours, about 1.5 hours, about 1 hour, about 50 minutes, about 40 minutes, about 30 minutes, about 20 minutes, about 10 minutes, about 5 minutes, about 2 minutes, about 1 minute, about 30 seconds or about 10 seconds to prevent, treat, ameliorate, mitigate, relieve, lighten, postpone, stop or halt one or more symptoms of T1D.
[00120] In some embodiments, a subject is orally administered one or more doses of a about 100 pg/kg or less, preferably about 95 pg/kg or less, 90 pg/kg or less, 85 pg/kg or less, 80 pg/kg or less, 75 pg/kg or less, 70 pg/kg or less, 65 pg/kg or less, 60 pg/kg or less, 55 pg/kg or less, 50 pg/kg or less, 45 pg/kg or less, 40 pg/kg or less, 35 pg/kg or less, 30 pg/kg or less, 25 pg/kg or less, 20 pg/kg or less, 15 pg/kg or less, 10 pg/kg or less, 5 pg/kg or less, 2.5 pg/kg or less, 2 pg/kg or less, 1.5 pg/kg or less, 1 pg/kg or less, 0.5 pg/kg or less, or 0.2 pg/kg or less of the anti- CD3 antibody such as teplizumab, otelixizumab or foralumab, to prevent, treat, ameliorate, mitigate, relieve, lighten, postpone, stop or halt one or more symptoms of T1D. In some embodiments, the oral dose of about 100 pg/kg or less, 95 pg/kg or less, 90 pg/kg or less, 85 pg/kg or less, 80 pg/kg or less, 75 pg/kg or less, 70 pg/kg or less, 65 pg/kg or less, 60 pg/kg or less, 55 pg/kg or less, 50 pg/kg or less, 45 pg/kg or less, 40 pg/kg or less, 35 pg/kg or less, 30 pg/kg or less, 25 pg/kg or less, 20 pg/kg or less, 15 pg/kg or less, 10 pg/kg or less, 5 pg/kg or less, 2.5 pg/kg or less, 2 pg/kg or less, 1.5 pg/kg or less, 1 pg/kg or less, 0.5 pg/kg or less, or 0.2 pg/kg or less of the anti-CD3 antibody such as teplizumab, otelixizumab or foralumab, is administered over about 6 hours, about 4 hours, about 2 hours, about 1.5 hours, about 1 hour, about 50 minutes, about 40 minutes, about 30 minutes, about 20 minutes, about 10 minutes, about 5 minutes, about 2 minutes, about 1 minute, about 30 seconds or about 10 seconds to prevent, treat, ameliorate, mitigate, relieve, lighten, postpone, stop or halt one or more symptoms of TID.
[00121] In some embodiments in which escalating doses are administered for the first days of the dosing regimen, the dose on day 1 of the regimen is about 5-150 pg/m2/day, preferably about 55-150 pg/m2/day, for example about 60-100 pg/m2/day and escalates to the daily dose as recited immediately above by day 3, 4, 5, 6 or 7. In some embodiments, on day 1, the subject is administered a dose of approximately 60 pg/m2/day, on day 2 approximately 125 pg/m2/day, on day 3 approximately 250 pg/m2/day, on day 4 approximately 500 pg/m2/day and on subsequent days of the regimen (e.g., days 5-14) 1,000 pg/m2/day. In some embodiments, on day 1, the subject is administered a dose of approximately 60 pg/m2/day, on day 2 approximately 125 pg/m2/day, on day 3 approximately 250 pg/m2/day, on day 4 approximately 500 pg/m2/day and on subsequent days of the regimen (e.g., days 5-14) 1,030 pg/m2/day. In some embodiments, on day 1, the subject is administered a dose of approximately 100 pg/m2/day, on day 2 approximately 425 pg/m2/day, on day 3 approximately 850 pg/m2/day, on day 4 approximately 850 ug/m2/day and on subsequent days of the regimen (e.g., days 5-14) 1,000 pg/m2/day. In some embodiments, on day 1, the subject is administered a dose of approximately 60 pg/m2/day, on day 2 approximately 125 pg/m2/day, on day 3 approximately 250 pg/m2/day, on day 4 approximately 500 pg/m2/day and on subsequent days of the regimen (e.g., days 5-14) 1,070 pg/m2/day.
[00122] In some embodiments, the initial dose is 1/4, to 1/2, to equal to the daily dose at the end of the regimen but is administered in portions at intervals of 6, 8, 10 or 12 hours. For example, a 13 pg/kg/day dose is administered in four doses of 3-4 pg/kg at intervals of 6 hours to reduce the level of cytokine release caused by administration of the antibody. In some embodiments, to reduce the possibility of cytokine release and other adverse effects, the first 1, 2, 3, or 4 doses or all the doses in the regimen are administered more slowly by intravenous administration. For example, a dose of 51 pg/m2/day may be administered over about 5 minutes, about 15 minutes, about 30 minutes, about 45 minutes, about 1 hour, about 2 hours, about 4 hours, about 6 hours, about 8 hours, about 10 hours, about 12 hours, about 14 hours, about 16 hours, about 18 hours, about 20 hours, and about 22 hours. In some embodiments, the dose is administered by slow infusion over a period of, e.g., 20 to 24 hours. In some embodiments, the dose is infused in a pump, preferably increasing the concentration of antibody administered as the infusion progresses.
[00123] In some embodiments, a set fraction of the doses for, for example, the 60 pg nr/day to 1000 pg/nr/dav or the 65 pg m2/day to 1030 pg/m2/day regimen described above is administered in escalating doses. In some embodiments, the fraction is 1/10, 1/4, 1/3, 1/2, 2/3 or 3/4 of the daily doses of the regimens described above.
[00124] In some embodiments, the anti-CD3 antibody such as teplizumab, otelixizumab or foralumab, is not administered by daily doses over a number of days, but is rather administered by infusion in an uninterrupted manner over a minimum of 30 minutes, for example 30 minutes, 1 hours, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 12 hours, 15 hours, 18 hours, 20 hours, 24 hours, 30 hours or 36 hours. The infusion may be constant or may start out at a lower dosage for, for example, the first 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours or 8 hours of the infusion and then increase to a higher dosage thereafter. Over the course of the infusion, the patient receives a dose equal to the amount administered in the 5 to 20-day regimens set forth above (for example the 14-day regimen set forth above). For example, a cumulative dose of approximately or greater than 9500 pg/m2, 10000 pg/m2, 10500 pg/m2, 11000 pg/m2, 11500 pg/m2, 12000 pg/m2, 12500 pg/m2, 13000 pg/m2, 13500 pg/m2 or 14000 pg/m2 is administered by infusion to the subject in need thereof. For example, a cumulative dose of teplizumab of approximately or greater than 11,240 pg/m2 is administered by infusion to the subject in need thereof over a period of at least 30 minutes. In particular, the speed and duration of the infusion is designed to minimize the level of free anti- CD3 antibody such as teplizumab, otelixizumab or foralumab in the subject after administration. In some embodiments, the level of free anti-CD3 antibody such as teplizumab should not exceed 200 ng/ml free antibody. In addition, the infusion is designed to achieve a combined T cell receptor coating and modulation of at least 50%, 60%, 70%, 80%, 90%, 95% or of 100%.
[00125] In some embodiments, the subject in need thereof is administered prior to the administering of the course IV infusion (for example the 14-day s course IV infusion) on each of days 1-5 an effective amount of pain reliever (such as a nonsteroidal anti-inflammatory drug (NSAID), acetaminophen), an antihistamine, an antiemetic or a combination thereof. In some embodiments, the pain reliever (such as a nonsteroidal anti-inflammatory drug (NSAID), acetaminophen), an antihistamine, an antiemetic or a combination thereof can be administered on each of days 1-5, 1-6, 1-7, 1-8, 1-9, 1-10, 1-11, 1-12, 1-13, 1-14 or during the duration of the treatment with the anti-CD3 antibodies. In some embodiments, the NSAID, acetaminophen, antihistamine, antiemetic or combination thereof is administered orally. In some embodiments, the NSAID, acetaminophen, antihistamine, antiemetic or combination thereof is administered intravenously. In some embodiments, antipyretics, antihistamines and/or antiemetics are administered to the subject in need thereof to mitigate cytokine release syndrome. In some embodiments, the liver enzymes are monitored and treatment with the anti-CD3 antibodies is discontinued or paused in subjects developing elevated ALT or AST more than 5 times the upper limit of normal.
[00126] In some embodiments, the anti-CD3 antibody such as teplizumab, otelixizumab or foralumab is administered chronically to treat, prevent, or slow or delay the onset or progression, or ameliorate, mitigate, relieve, lighten, postpone, stop or halt one or more symptoms of type 1 diabetes. For example, in some embodiments, a low dose of the anti-CD3 antibody such as teplizumab is administered once a month, twice a month, three times per month, once a week or even more frequently either as an alternative to the 6 to I4-day dosage regimen discussed above or after administration of such a regimen to enhance or maintain its effect. Such a low dose may be anywhere from about 1 pg/m2 to about 100 pg/m2. such as approximately 5 pg/m2. 10 pg/m2, 15 pg/m2, 20 pg/m2, 25 pg/m2, 30 pg/m2, 35 pg/m2, 40 pg/m2, 45 pg/m2, 50 pg/m2, 55 pg/m2, 60 pg/m2, 65 pg/m2, 70 pg/m2, 75 pg/m2, 80 pg/m2, 85 pg/m2, 90 pg/m2, 95 pg/m2 or 100 pg/m2.
[00127] In some embodiments, the anti-CD3 antibody such as teplizumab, otelixizumab or foralumab is administered by infusion in medical facilities, outpatient infusion centers. Yet in other embodiments, the anti-CD3 antibody such as teplizumab, otelixizumab or foralumab is administered by infusion in a home setting. Home infusion therapy involves the administration of the therapeutic agents, for example, anti-CD3 antibody using intravenous, or subcutaneous routes, in the patient's home rather than in a physician's office or hospital. Infusion therapies in the home can be administered by a home health care worker or by a patient himself. In some embodiments, a health care worker having some training in the operation of infusion equipment and the administration of anti-CD3 antibody can provide the patient with self-administration training and all the necessary equipment and/or supplies needed for the administration.
[00128] In some embodiments, the subject may be re-dosed at some time subsequent to administration of the anti-CD3 antibody such as teplizumab, otelixizumab or foralumab dosing regimen, for example, based upon one or more physiological parameters or may be done as a mater of course. Such redosing may be administered and/or the need for such redosing evaluated 2 months, 4 months, 6 months, 8 months, 9 months, 1 year, 15 months, 18 months, 2 years, 30 months or 3 years after administration of a dosing regimen and may include administering a course of treatment every 6 months, 9 months, 1 year, 15 months, 18 months, 2 years, 30 months or 3 years indefinitely.
[00129] Some embodiments relate to an anti-CD3 antibody for use in a method of preventing or delaying the onset of clinical type 1 diabetes (T1D), comprising administering a prophy lactically effective amount of an anti-CD3 antibody to a non-diabetic subject who is at risk of T1D, wherein the prophylactically effective amount has a cumulative dose of about 10,500 pg/m2 to about 14,000 pg/m2; and determining, prior to or after the administering step, that the non-diabetic subject has more than about 10% TIGIT+KLRG1+CD8+ T-cells in all CD3+ T cells, which is indicative of successful prevention or delay of the onset of clinical T1D.
[00130] Aspects of the disclosure relate to a method of preventing or delaying onset of clinical type 1 diabetes (T1D), comprising administering a prophylactically effective amount of an anti- CD3 antibody to a non-diabetic subject who is at risk of T1D, wherein the prophylactically effective amount has a cumulative dose of from about 10,500 pg/m2 to about 14,000 pg/m2.
[00131] In some embodiments, the subject is 8 years of age or older. In some embodiments, the subject is 5 years of age or older. In some embodiments, the subject is 3 years of age or older. In some embodiments, the subject is 1 year of age or older. In some embodiments, the subject is 1 year old or younger.
[00132] In some embodiments, the non-diabetic subject is a relative of a patient with T1D.
[00133] In some embodiments, the method further comprises determining that the non-diabetic subject (1) is negative for zinc transporter 8 (ZnT8) antibodies, (2) is HLA-DR4+, and/or (3) is not HLA-DR3+. In some embodiments, the non-diabetic subject does not have antibodies against ZnT8. In some embodiments, the non-diabetic subject is HLA-DR4+ and is not HLA-DR3+.
[00134] In some embodiments, the non-diabetic subject has 2 or more diabetes-related autoantibodies selected from islet cell antibodies (ICA), insulin autoantibodies (IAA), and antibodies to glutamic acid decarboxylase (GAD), tyrosine phosphatase (IA-2/ICA512) or ZnT8.
[00135] In some embodiments, the non-diabetic subject has abnormal glucose tolerance on oral glucose tolerance test (OGTT). In some embodiments, the abnormal glucose tolerance on OGTT is a fasting glucose level of 110-125 mg/dL, or 2 hour plasma of > 140 and < 200 mg/dL, or an intervening glucose value at 30, 60, or 90 minutes on OGTT > 200 mg/dL. [00136] In some embodiments, the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab. In some embodiments, the anti-CD3 antibody is teplizumab.
[00137] In some embodiments, the prophy tactically effective amount comprises a 14-day course of subcutaneous (SC)inj ection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody with a cumulative dose of from about 11,000 pg/m2 to about 14,000 pg/m2.
[00138] In some embodiments, the method comprises administering a 14-day course IV infusion at about 60 pg/m2 on day 1, about 125 pg/m2 on day 2, about 250 pg/m2 on day 3, and about 500 pg/m2 on day 4, and a dose of about 1,000 pg/m2 on each of days 5-14.
[00139] In some embodiments, the method comprises administering a 14-day course IV infusion at about 60 pg/m2, about 125 pg/m2 on day 1, about 250 pg/m2 on day 2, and about 500 pg/m2 on day 3, and a dose of about 1,030 pg/m2 on each of days 5-14.
[00140] In some embodiments, the method comprises administering a 14-day course IV infusion at about 100 pg/m2 on day 1, about 425 pg/m2 on day 2, about 850 pg/m2 on day 3, about 850 pg/m2 on day 4, and a dose of about 1,000 pg/m2 on each of days 5-14.
[00141] In some embodiments, the method comprises administering a 14-day course IV infusion at about 65 pg/m2 on day 1, about 125 pg/m2 on day 2, about 250 pg/m2 on day 3, and about 500 pg/m2 on day 4, and a dose of about 1,070 pg/m2 on each of days 5-14.
[00142] In some embodiments, the method comprises administering a 14-day course IV infusion at about 65 pg/m2 on day 1, about 125 pg/m2 on day 2, about 250 pg/m2 on day 3, and about 500 pg/m2 on day 4, and a dose of about 1,030 pg/m2 on each of days 5-14.
[00143] In some embodiments, the prophylactically effective amount delays median time to clinical diagnosis of T1D by at least 50%, at least 80%, or at least 90%. In some embodiments, the prophylactically effective amount delays median time to clinical diagnosis of T1D by at least 12 months, at least 18 months, at least 24 months, at least 36 months, at least 48 months, or at least 60 months.
[00144] In some embodiments, the method further comprises determining, prior to or after the administering step, that the non-diabetic subject has more than about 10% TIGIT+KLRG1+CD8+ T-cells in all CD3+ T cells, which is indicative of successful prevention or delay of the onset of clinical T1D. In some embodiments, the determining of TIGIT+KLRG1+CD8+ T-cells is by flow cytometry. In some embodiments, the method further comprises determining a decrease in a percentage of CD8+ T cells expressing proliferation markers Ki67 and/or CD57.
[00145] Aspects of the disclosure relate to a method of preventing or delaying onset of clinical type 1 diabetes (T1D), comprising administering to a non-diabetic subject 8 years of age or older who is at risk of T1D a 14-day course IV infusion of teplizumab at about 65 pg/m2 on day 1, about 125 pg/m2 on day 2, about 250 pg/m2 on day 3, and about 500 pg/m2 on day 4, and a dose of about 1,030 pg/m2 on each of days 5-14.
[00146] Aspects of the disclosure relate to a method of delaying onset of Stage 3 type 1 diabetes (T1D), comprising administering to a subject in need thereof who has a diagnosis of stage 2 T1D a 14-day course IV infusion of teplizumab at about 65 pg/m2 on day 1, about 125 pg/m2 on day 2, about 250 pg/m2 on day 3, and about 500 pg/m2 on day 4, and a dose of about 1,030 pg/m2 on each of days 5-14.
[00147] In some embodiments, the subject in need thereof is an adult. In some embodiments, the subject in need thereof is a pediatric subject 8 years of age or older. In some embodiments, the subject in need thereof is a pediatric subject 5 years of age or older. In some embodiments, the subject in need thereof is a pediatric subject 3 years of age or older. In some embodiments, the subject in need thereof is a pediatric subject 1 year of age or older. In some embodiments, the subject in need thereof is a pediatric subject 1 year old or younger.
[00148] In some embodiments, the method comprises documenting at least two positive pancreatic islet autoantibodies in the subject who has dysglycemia without overt hyperglycemia before administering the 14-day course. In some embodiments, the subject in need thereof has dysglycemia without overt hyperglycemia and has two or more pancreatic islet autoantibodies. In some embodiments, the two or more pancreatic islet autoantibodies comprise islet cell antibodies (ICA), insulin autoantibodies (IAA), and antibodies to glutamic acid decarboxylase (GAD), tyrosine phosphatase (IA-2/ICA512) or ZnT8. In some embodiments, the method comprises administering on at least each of days 1-5, and prior to the administering of the 14- day course IV infusion, an effective amount of a nonsteroidal anti-inflammatory drug (NSAID), acetaminophen, an antihistamine, an anti emetic or a combination thereof. In some embodiments, the method comprises administering orally the NSAID, acetaminophen, antihistamine, antiemetic or combination thereof. In some embodiments, the method comprises administering the 14-day course IV infusion once daily for 14 consecutive days over a period of at least 30 minutes. [00149] Aspects of the disclosure relate to a method of prognosing responsiveness of an anti- CD3 antibody in preventing or delaying onset of type 1 diabetes (T1D), the method comprising administering a prophylactically effective amount of an anti-CD3 antibody to a non-diabetic subject who is at risk of T1D, wherein the prophylactically effective amount has a cumulative dose of about 10,500 pg/m2 to about 14,000 pg/m2; and determining C-peptide area under the curve (AUC): glucose AUC ratio, wherein an increase in the ratio indicates responsiveness to the anti-CD3 antibody and/or non-progression to clinical T1D.
[00150] Aspects of the disclosure relate to an anti-CD3 antibody for use in a method of preventing or delaying onset of clinical type 1 diabetes (T1D), comprising: administering a prophylactically effective amount of an anti-CD3 antibody to a non-diabetic subject who is at risk of T1D, wherein the prophylactically effective amount has a cumulative dose of about 10,500 pg/m2 to about 14,000 pg/m2.
[00151] Aspects of the disclosure relate to a teplizumab composition for use in a method of delaying onset of Stage 3 type 1 diabetes (T1D), comprising administering to a subject in need thereof who has a diagnosis of stage 2 T1D a 14-day course IV infusion at about 65 pg/m2 on day 1, about 125 pg/m2 on day 2, about 250 pg/m2 on day 3, and about 500 pg/m2 on day 4, and a dose of about 1,030 pg/m2 on each of days 5-14.
EXAMPLES
Example 1:
[00152] Type 1 diabetes (T1D) is an autoimmune disease characterized by T-cell mediated destruction of insulin producing beta cells within the pancreatic islets of Langerhans. Longitudinal observational studies over more than 30 years have described the progression of the autoimmune disease from the first appearance of autoantibodies until beta cell function is critically impaired and the clinical diagnosis, often with ketoacidosis, occurs (7-5). T1D is associated with a need for lifelong exogenous insulin administration for survival, increased morbidity and mortality due to immediate (e.g. hypoglycemia) and long-term complications (e.g. vascular, renal, and eye disease), and reduced life-span, life impairments, and considerable health-care-related costs (6-9). Thus, approaches to prevent progression to clinical T1D before irremediable beta cell destruction and insulin deficiency, are of paramount importance.
[00153] Changes in beta cell function precede the clinical diagnosis of T1 D and have been studied in natural history cohorts of individuals who are identified as at-risk for the disease based on the presence of islet autoantibodies (10-12). Some studies suggest an ongoing and intermittently progressive decline in beta cell function, that begins years before clinical diagnosis at a time when glucose tolerance is normal. During this period there are signs of ongoing autoimmunity: Based on the findings of the natural history, individuals with two or more islet autoantibodies have been classified as stages of T1D, with further specification according to the level of metabolic dy sfunction: Stage 1 prior to glucose abnormalities, Stage 2 with dysglycemia during an oral glucose tolerance test (OGTT), and Stage 3 at clinical presentation with hyperglycemia (2, 13, 14). However, the relationships between changes in beta cell function and clinical disease remain poorly defined. It is known, for example, that glucose tolerance, defined through responses to an oral glucose tolerance test (OGTT), may fluctuate between abnormal and normal values within an individual who is at risk (15, 16). In addition, OGTT glucose tolerance classifications used to designate a clinical diagnosis, and beta cell function, measured by C-peptide responses to a metabolic challenge, may not be closely related and at diagnosis using an OGTT, many individuals have clinically meaningful C-peptide responses (15-18).
[00154] The following are dosing regimens matching AUCinf, Cmax and Ctroughis (trough concentration prior to Day 14 dosing) exposure parameters of the reference regimen (Herold 14- day regimen used in Protege and TN- 10 studies, Herold et al NEJM 2019).
[00155] Individual (conditional) PK simulations were performed using Provention Bio Model 363 (Regimen A, B and C), Model 372 (Regimen D) and individual model parameters from the corresponding arms of the PROTECT Phase 3 study of teplizumab in newly-diagnosed T1D patients.
[00156] Typical (average) simulations for comparison of exposure values were also performed.
[00157] More than 30 regimens based on the Herold 14-day regimen or PROTECT regimen were simulated and were compared to the reference regimen. Alternative dosage regimens are presented in Table 1.
Table 1 Regimens Used in Clinical Studies and Proposed Alternative Regimens for anti-CD3 antibody
Figure imgf000041_0001
Figure imgf000042_0001
Dosing in pg/m2 via 30-minute infusion
[00158] Each of these regimens includes a 4-day ramp-up followed by 10 consecutive days of maintenance dosing, with an infusion duration of 30 minutes. As in previous T1D regimens, the ramp-up is important to avoid the occurrence of acute events such as those related to cytokine release.
• Regimen A: Incorporates the backbone of the TN-10 (Herold 14-day) 4-day ramp-up and maintenance doses, with an approximate 18-20% upward adjustment for each daily dose. This regimen meets the bioequivalence criteria for the three PK parameters under both conditional and typical (average) simulations with the exception that the average (typical) simulation ratio for CtroughB is slightly less than 0.80 (0.779). (Table 2). The cumulative dose for this regimen is 10,935 pg/m2 (approximately 21% higher compared to the Herold 14-day regimen). o Day 1: 60 pg/m2 o Day 2: 125 pg/m2 o Day 3: 250 pg/m2 o Day 4: 500 pg/m2 o Days 5-14: 1000 pg/m2 • Regimen B: Incorporates the backbone of the TN-10 (Herold 14-day) regimen and an approximate 18-25% upward adjustment for each daily dose; this has a similar 4-day ramp- up as in Regimen A, but the maintenance dose is 1030 pg/m2. This regimen meets all bioequivalence criteria for AUCinf, Cmax and Ctroughis under both the conditional and typical (average) simulations (Table 2). The cumulative dose for this regimen is 11,235 pg/m2 (approximately 24% higher compared to the Herold 14-day regimen). o Day 1: 60 pg/m2 o Day 2: 125 pg/m2 o Day 3: 250 pg/m2 o Day 4: 500 pg/m2 o Days 5-14: 1030 pg/m2
• Regimen D: Incorporates the backbone of the TN-10 (Herold 14-day) regimen and an approximate 18-25% upward adjustment for each daily dose; this has a similar 4-day ramp- up as in Regimen A, but the Day 1 dose is 65 pg/m2 and the maintenance dose is 1030 pg/m2. This regimen meets all bioequivalence criteria for AUCinf, Cmax and Ctroughi3 under both the conditional and typical (average) simulations (Table 2). The cumulative dose for this regimen is 11,240 pg/m2 (approximately 24% higher compared to the Herold 14-day regimen). o Day 1: 65 pg/m2 o Day 2: 125 pg/m2 o Day 3: 250 pg/m2 o Day 4: 500 pg/m2 o Days 5-14: 1030 pg/m2
• Regimen C : Incorporates the backbone of the PROTECT 12-day regimen with a 2-day ramp- up, with minor adjustment for the Day 1 dose from 106 pg/m2 to 100 pg/m2 to simplify dose calculations and preparations; the two doses of 850 pg/m2 on Day 3 and Day 4 replicate the PROTECT regimen before the administration of the maintenance dose of 1000 pg/m2. This regimen meets 80-125% bioequivalence criteria for AUCinf, Cmax and Ctroughia under both the conditional and typical (average) simulations (Table 2). The cumulative dose for this regimen is 12,225 pg/m2 (approximately 35% higher compared to the Herold 14-day regimen). o Day 1: 100 pg/m2 o Day 2: 425 pg/m2 o Day 3: 850 pg/m2 o Day 4: 850 pg/m2 o Days 5-14: 1000 pg/m2
Table 2 Comparison of Alternative Regimens to Reference Regimen: Conditional and Typical (average) Simulations
Figure imgf000044_0001
Typical simulations performed using median BSA and weight of subjects in the PROTECT study and no anti-drug antibodies (ADA)
[00159] A comparison of the distribution of exposures (AUCinf, Cmax and Ctroughts) for the reference regimen (Herold 14-day) and the anti-CD3 antibody exposures predicted for the alternative regimens are shown in FIG. 1 and FIG. 5. A comparison of the distribution of exposures (AUCinf, Cmax and Ctroughis) for the Protege and TN-10 regimen (Herold 14-day) and the anti-CD3 antibody exposures predicted for the alternative regimens are shown in FIG. 2 and FIG. 6. Safety considerations
[00160] These regimens are predicted not to result in additional safety findings. The predicted Cmax and Ctrough during each day of the 14-day course for the alternative regimens are shown in FIG. 3 (Regimens A and B), FIG. 4 (Regimen C) and FIG. 7 (Regimen D) alongside the values from the historical clinical trials. The Cmax and Ctrough values for Regimens A and B do not exceed those observed in Protege or TN 10 using the Herold 14-day regimen. The Cmax and Ctrough values for Regimen C do not exceed those observed in the PROTECT 12-day regimen.
[00161] Additionally, we evaluated the safety profile of patients in the Protege and TN-10 studies who have AUCinr greater than and less than or equal to 6000 ng*day/mL. This value represents approximately the geometric mean exposure for the proposed alternative dosing regimens (Table 3). As shown in Table 4 the safety profile based on AEs reported in at least 5% of patients appear comparable between the subset of patients w ith AUC >6000 ng*day/mL and those with <6000 ng*day/mL.
Table 3 Simulated AUCinr for Alternative Regimens for the anti-CD3 antibody
Figure imgf000045_0001
Table 4 Safety Profile for Patients with AUCinf >6000 ng*day/mL and with <6000 ng*day/mL: AEs Reported in >5% of Patients
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
[00162] Safety information for the precursor product to teplizumab, hOKT3yl (Ala-Ala), is available for non-TID patients, including renal transplant (Woodie 1999), islet transplant (Hering 2004) and psoriatic arthritis (Utset 2002) patients. Total cumulative doses evaluated over a 10 consecutive day treatment period included 76 mg, 46 mg and 40 mg for renal transplant, islet transplant and psoriatic arthritis patients, respectively. The safety events were similar to those observed in T1D patients who received an average total cumulative dose of ~18- 20 mg over a 12-14-day treatment period.
Example 2: Additional dosage regimens
[00163] Alternative dosage regimens are shown in the Table 5 below:
Table 5: Simulated Alternative 14-days Regimens
Figure imgf000049_0002
Figure imgf000050_0001
Example 3: Additional dosage regimens
[00164] Alternative dosage regimen G is show n in the Table 6 below:
Table 6:
Figure imgf000050_0002
Example 4: Dosage and Administration [00165] The teplizumab compositions and methods provided herein are indicated to delay the onset of Stage 3 type 1 diabetes in adults and pediatric patients 8 years of age and older with Stage 2 type 1 diabetes.
[00166] Teplizumab-mzwv (also referred herein as Teplizumab) is a CD3-directed monoclonal antibody (humanized IgGl kappa) that has a molecular weight of approximately 150 kilodalton (kDa) and is expressed from a recombinant Chinese hamster ovary (CHO) cell line.
[00167] Teplizumab injection is supplied as a sterile, preservative-free, clear and colorless solution in a 2 mg/2 mL (1 mg/mL) single-dose vial for intravenous use. Each mL contains 1 mg of teplizumab-mzwv, dibasic sodium phosphate (0.26 mg), monobasic sodium phosphate (0.98 mg), polysorbate 80 (0.05 mg), sodium chloride (8.78 mg), and water for injection. The pH is 6.1.
CLINICAL PHARMACOLOGY
[00168] Mechanism of Action
[00169] Teplizumab-mzwv binds to CD3 (a cell surface antigen present on T lymphocytes) and delays the onset of Stage 3 type 1 diabetes in adults and pediatric patients aged 8 years and older with Stage 2 type 1 diabetes. The mechanism may involve partial agonistic signaling and deactivation of pancreatic beta cell auto-reactive T lymphocytes. Teplizumab-mzwv leads to an increase in the proportion of regulatory T cells and of exhausted CD8+ T cells in peripheral blood.
[00170] Pharmacodynamics
[00171] Clinical studies have shown that teplizumab-mzwv binds to CD3 molecules on the surface of both CD4+ and CD8+ T cells during treatment, with internalization of the teplizumab- mzwv/CD3 complex from the surface of T cells. Pharmacodynamic effects include lymphopenia in the absence of depletion of T cells with a nadir on the 5th day of dosing, during a 14-day course of teplizumab-mzwv treatment. Teplizumab-mzwv exposure-response relationship and time course of pharmacodynamic response for the safety and effectiveness of teplizumab-mzwv have not been fully characterized.
[00172] Pharmacokinetics
[00173] Steady state concentrations of teplizumab-mzwv are not expected to be achieved during the 14-day course of teplizumab. [00174] Distribution
[00175] The central volume of distribution (Vd) of teplizumab-mzwv was 2.27 L in a 60 kg subject.
[00176] Elimination
[00177] Teplizumab-mzwv showed saturable binding and elimination. The mean (SD) terminal elimination half-life and clearance of teplizumab-mzwv are 4.5 (0.2) days and 2.7 (0.8) L/day in a 60 kg subject, respectively.
[00178] Metabolism
[00179] Teplizumab-mzwv is expected to be metabolized into small peptides by catabolic pathways.
[00180] Specific Populations
[00181] No clinically significant differences in the pharmacokinetics of teplizumab-mzwv were observed based on age (8 to 35 years old), biologic sex or racial groups (White, Asians).
[00182] Body Weight
[00183] BSA-based dosing normalizes the exposure to teplizumab-mzwv across body weight.
[00184] Immunogenicity
[00185] The observed incidence of anti-drug antibodies is highly dependent on the sensitivity and specificity of the assay. Differences in assay methods preclude meaningful comparisons of the incidence of anti-drug antibodies in the studies described below with the incidence of antidrug antibodies in other studies, including those of teplizumab-mzwv or of other teplizumab products.
[00186] In the placebo- controlled study in patients aged 8 years of age and older with Stage 2 type 1 diabetes (Study TN-10), approximately 59% of teplizumab-treated patients developed anti-teplizumab-mzwv antibodies, 46% of whom developed neutralizing antibodies. There is insufficient information to characterize the effects of ADA on pharmacokinetics, pharmacodynamics or effectiveness of teplizumab-mzwv. There was a higher incidence of rash in teplizumab-treated patients who developed anti-tephzumab-mzwv antibodies compared to those who did not develop anti-tephzumab-mzwv antibodies.
[00187] Patient Selection [00188] Select adult patients and pediatric patients 8 years of age and older for TZIELD treatment who have a diagnosis of Stage 2 type 1 diabetes.
[00189] Confirm Stage 2 type 1 diabetes by documenting at least two positive pancreatic islet cell autoantibodies (FDA-authorized tests for pancreatic islet cell autoantibodies are recommended) in those who have dysglycemia without overt hyperglycemia. In patients who meet criteria for a diagnosis of Stage 2 type 1 diabetes, ensure the clinical history of the patient does not suggest type 2 diabetes.
[00190] Administration Instructions and Recommended Dosage
[00191] Dilute prior to use:
• Each vial is intended for single-dose only. Prepare a: Sterile glass vial with 18 mL of 0.9% Sodium Chloride Injection or Polyvinylchloride (PVC) infusion bag with 18 mL of 0.9% Sodium Chloride Injection.
• Remove 2 mL of teplizumab from the vial and slowly add to the 18 mL of 0.9% Sodium Chloride Injection. Mix gently by slowly inverting the vial or rocking the infusion bag. The resulting 20 mL diluted solution contains 100 mcg/mL of teplizumab-mzwv.
• Using an appropriately sized syringe (e g., 5 mL), withdraw the volume of diluted solution required on that day for the calculated dose from the 100 mcg/mL solution.
• Slowly add contents of the syringe containing teplizumab dose to a 25 mL 0.9% sodium chloride injection PVC infusion bag. Gently rock the infusion bag to ensure that the solution mixes sufficiently. Do not shake.
• Discard unused portion of remaining diluted teplizumab solution in the sterile glass vial or PVC infusion bag.
• Start the infusion of teplizumab within 2 hours of preparation. If not used immediately, store the diluted solution at room temperature [15°C to 30°C (59°F to 86°F)] and complete infusion within 4 hours of the start of preparation. Discard the diluted solution if not administered within 4 hours of preparation.
[00192] Premedicate prior to infusion for the first 5 days of dosing with oral medications including: (1) a nonsteroidal anti-inflammatory drug (NSAID) or acetaminophen, (2) an antihistamine, and/or (3) an antiemetic. Administer additional doses of premedication if needed.
[00193] Administer by intravenous infusion (over a minimum of 30 minutes) once daily for 14 consecutive days. [00194] The recommended dosage for pediatrics and adult patients ages 8 years and older, based on body surface area (BSA) is described in the Table below.
[00195] Do not administer two doses on the same day.
Table: Recommended Once Daily Dosage in Adults and Pediatric Patients 8 Years of Age and Older During the 14-Day Treatment (Regimen D)
Figure imgf000054_0001
Recommendations Regarding Missed Dose(s)
[00196] If a planned infusion is missed, resume dosing by administering all remaining doses on consecutive days to complete the 14-day treatment course.
Laboratory Evaluation, and Vaccination Prior to Initiation
[00197] Prior to initiating, obtain a complete blood count and liver enzy me tests.
[00198] Use of is not recommended in patients with:
Lymphocyte count less than 1,000 lymphocytes/mcL
Hemoglobin less than 10 g/dL
Platelet count less than 150,000 platelets/mcL
Absolute neutrophil count less than 1500 neutrophils/mcL
Elevated ALT or AST greater than 2 times the upper limit of normal (ULN) or bilirubin greater than 1.5 times ULN
Laboratory or clinical evidence of acute infection with Epstein-Barr virus (EBV) or cytomegalovirus (CMV)
Active serious infection or chronic active infection other than localized skin infections
Administer all age-appropriate vaccinations prior to starting administration
Administer live-attenuated (live) vaccines at least 8 weeks prior to treatment.
Administer inactivated (killed) vaccines or mRNA vaccines at least 2 weeks prior to treatment.
Dosage Forms and Strengths [00199] Injection: 2 mg per 2 mL (1 mg/mL) clear and colorless solution in a single-dose vial.
Contraindications
[00200] None.
[00201] Modifications and variations of the described methods and compositions of the present disclosure will be apparent to those skilled in the art without departing from the scope and spirit of the disclosure. Although the disclosure has been described in connection with specific embodiments, it should be understood that the disclosure as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the disclosure are intended and understood by those skilled in the relevant field in which this disclosure resides to be within the scope of the disclosure as represented by the following claims.
INCORPORATION BY REFERENCE
[00202] All patents and publications mentioned in this specification are herein incorporated by reference to the same extent as if each independent patent and publication was specifically and individually indicated to be incorporated by reference.
REFERENCES
1. A. G. Ziegler, M. Rewers, O. Simell, T. Simell, J. Lempainen, A. Steck, C. Winkler, J. Ilonen, R. Veijola, M. Knip, Seroconversion to multiple islet autoantibodies and risk of progression to diabetes in children. Jama 309, 2473-2479 (2013).
2. R. A. Insel, J. L. Dunne, M. A. Atkinson, J. L. Chiang, D. Dabelea, P. A. Gottlieb, C. J. Greenbaum, K. C. Herold, J. P. Krischer, A. Lemmark, R. E. Ratner, M. J. Rewers, D. A. Schatz, J. S. Skyler, J. M. Sosenko, A. G. Ziegler, Staging presy mptomatic type 1 diabetes: a scientific statement of JDRF, the Endocrine Society, and the American Diabetes Association. Diabetes Care 38, 1964-1974 (2015).
3. J. P. Krischer, K. F. Lynch, A. Lemmark, W. A. Hagopian, M. J. Rewers, J. X. She, J. Toppan, A. G. Ziegler, B. Akolkar, T. S. Group, Genetic and Environmental Interactions Modify the Risk of Diabetes-Related Autoimmunity by 6 Years of Age: The TEDDY Study. Diabetes Care 40, 1194-1202 (2017).
4. J. L. Mahon, J. M. Sosenko, L. Rafkin-Mervis, H. Krause-Steinrauf, J. M. Lachin, C. Thompson, P. J. Bingley, E. Bonifacio, J. P. Palmer, G. S. Eisenbarth, J. Wolfsdorf, J. S. Skyler,
C. TrialNet Natural History, G. Type 1 Diabetes TrialNet Study, The TrialNet Natural History Study of the Development of Type 1 Diabetes: objectives, design, and initial results. Pediatr Diabetes 10, 97-104 (2009).
5. H. T. Siljander, R. Hermann, A. Hekkala, J. Lahde, L. Tanner, P. Keskinen, J. Ilonen, O. Simell, R. Veijola, M. Knip, Insulin secretion and sensitivity in the prediction of type 1 diabetes in children with advanced beta-cell autoimmunity. Eur J Endocrinol 169, 479- 503 (2013).
6. N. C. Foster, R. W. Beck, K. M. Miller, M. A. Clements, M. R. Rickels, L. A. DiMeglio,
D. M. Maahs, W. V. Tamborlane, R. Bergenstal, E. Smith, B. A. Olson, S. K. Garg, State of Type 1 Diabetes Management and Outcomes from the T1D Exchange in 2016-2018. Diabetes Technol Ther 21, 66-72 (2019).
7. A. Rawshani, N. Sattar, S. Franzen, A. Rawshani, A. T. Hattersley, A. M. Svensson, B. Eliasson, S. Gudbjomsdottir, Excess mortality and cardiovascular disease in young adults with type 1 diabetes in relation to age at onset: a nationwide, register-based cohort study. Lancet 392, 477-486 (2018).
8. S. J. Livingstone, D. Levin, H. C. Looker, R. S. Lindsay, S. H. Wild, N. Joss, G. Leese, P. Leslie, R. J. McCrimmon, W. Metcalfe, J. A. McKnight, A. D. Morris, D. W. Pearson, J. R. Petrie, S. Philip, N. A. Sattar, J. P. Traynor, H. M. Colhoun, g. Scottish Diabetes Research Network epidemiology, R. Scottish Renal, Estimated life expectancy in a Scottish cohort with type 1 diabetes, 2008-2010. JAMA 313, 37-44 (2015).
9. B. Tao, M. Pietropaolo, M. Atkinson, D. Schatz, D. Taylor, Estimating the cost of type 1 diabetes in the U.S.: a propensity score matching method. PLoS One 5, el l501 (2010).
10. C. Evans-Molina, E. K. Sims, L. A. DiMeglio, El. M. Ismail, A. K. Steck, J. P. Palmer, J. P. Krischer, S. Geyer, P. Xu, J. M. Sosenko, G. Type 1 Diabetes TrialNet Study, beta Cell dysfunction exists more than 5 years before type 1 diabetes diagnosis. JCI InsightS' , (2018).
11. E. K Sims, L. A. DiMeglio, Cause or effect? A review of clinical data demonstrating beta cell dysfunction prior to the clinical onset of type 1 diabetes. Mol Metab 27S, S129-S138 (2019).
12. M. K. Koskinen, O. Helminen, J. Matomaki, S. Aspholm, J. Mykkanen, M. Makinen, V. Simell, M. Vaha-Makila, T. Simell, J. Ilonen, Reduced 0-cell function in early preclinical type 1 diabetes. European Journal of Endocrinology 174, 251-259 (2016).
13. P. J. Bingley, D. K. Wherrett, A. Shultz, L. E. Rafkin, M. A. Atkinson, C. J. Greenbaum, Type 1 Diabetes TrialNet: A Multifaceted Approach to Bringing Disease-Modifying Therapy to Clinical Use in Type 1 Diabetes. Diabetes Care 41, 653-661 (2018).
14. C. J. Greenbaum, C. Speake, J. Krischer, J. Buckner, P. A. Gottlieb, D. A. Schatz, K. C. Herold, M. A. Atkinson, Strength in Numbers: Opportunities for Enhancing the Development of Effective Treatments for Type 1 Diabetes-The TrialNet Experience. Diabetes 67, 1216-1225 (2018).
15. B. M. Nathan, D. Boulware, S. Geyer, M. A. Atkinson, P. Colman, R. Goland, W. Russell, J. M. Wentworth, D. M. Wilson, C. Evans-Molina, D. Wherrett, J. S. Skyler, A. Moran, J. M. Sosenko, T. Type 1 Diabetes, G. Diabetes Prevention Trial-Type 1 Study, Dysglycemia and Index60 as Prediagnostic End Points for Type 1 Diabetes Prevention Trials. Diabetes Care 40, 1494-1499 (2017).
16. J. M. Sosenko, J. P. Palmer, L. Rafkin-Mervis, J. P. Krischer, D. Cuthbertson, J. Mahon, C. J. Greenbaum, C. C. Cowie, J. S. Skyler, G. Diabetes Prevention Trial-Type 1 Study, Incident dysglycemia and progression to type 1 diabetes among participants in the Diabetes Prevention Trial-Type 1. Diabetes Care 32, 1603-1607 (2009).
17. J. P. Palmer, C-peptide in the natural history of type 1 diabetes. Diabetes Metab Res Rev 25, 325-328 (2009).
18. C. J. Greenbaum, A. M. Anderson, L. M. Dolan, E. J. Mayer-Davis, D. Dabelea, G. Imperatore, S. Marcovina, C. Pihoker, S. S. Group, Preservation of beta-cell function in autoantibody -positive youth with diabetes. Diabetes Care 32, 1839-1844 (2009).
19. W. Hagopian, R. J. Ferry, Jr., N. Sherry, D. Carlin, E. Bonvini, S. Johnson, K. E. Stein, S. Koenig, A. G. Daifotis, K. C. Herold, J. Ludvigsson, I. Protege Trial, Teplizumab preserves C-peptide in recent-onset type 1 diabetes: two-year results from the randomized, placebo- controlled Protege trial. Diabetes 62, 3901 -3908 (2013).
20. K. C. Herold, S. E. Gitelman, M. R. Ehlers, P. A. Gottlieb, C. J. Greenbaum, W. Hagopian, K. D. Boyle, L. Keyes-Elstein, S. Aggarwal, D. Phippard, P. H. Sayre, J. McNamara, J. A. Bluestone, A. T. E. S. T. Ab, Teplizumab (anti-CD3 mAb) treatment preserves C-peptide responses in patients with new-onset type 1 diabetes in a randomized controlled trial: metabolic and immunologic features at baseline identify a subgroup of responders. Diabetes 62. 3766-3774 (2013).
21. K. C. Herold, W. Hagopian, J. A. Auger, E. Poumian-Ruiz, L. Taylor, D. Donaldson, S. E. Gitelman, D. M. Harlan, D. Xu, R. A. Zivin, J. A. Bluestone, Anti-CD3 monoclonal antibody in new-onset type 1 diabetes mellitus. N Engl J Med 346, 1692-1698. (2002).
22. B. Keymeulen, E. Vandemeulebroucke, A. G. Ziegler, C. Mathieu, L. Kaufman, G. Hale, F. Gorus, M. Goldman, M. Walter, S. Candon, L. Schandene, L. Crenier, C. De Block, J. M. Seigneurin, P. De Pauw, D. Pierard, I. Weets, P. Rebello, P. Bird, E. Bertie, M. Frewin, H. Waldmann, J. F. Bach, D. Pipeleers, L. Chatenoud, Insulin needs after CD3 -antibody therapy in new-onset type 1 diabetes. N Engl J Med 352, 2598-2608 (2005).
23. S. A. Long, J. Thorpe, H. A. DeBerg, V. Gersuk, J. Eddy, K. M. Harris, M. Ehlers, K. C. Herold, G. T. Nepom, P. S. Linsley, Partial exhaustion of CD8 T cells and clinical response to teplizumab in new-onset type 1 diabetes. Sci Immunol 1, (2016).
24. N. Sherry, W. Hagopian, J. Ludvigsson, S. M. Jain, J. Wahlen, R. J. Ferry, Jr., B. Bode, S. Aronoff, C. Holland, D. Carlin, K. L. King, R. L. Wilder, S. Pillemer, E. Bonvini, S. Johnson, K. E. Stein, S. Koenig, K. C. Herold, A. G. Daifotis, I. Protege Trial, Teplizumab for treatment of type 1 diabetes (Protege study): 1-year results from a randomised, placebo-controlled trial. Lancet 378, 487-497 (2011).
25. J. E. Tooley, N. Vudattu, J. Choi, C. Cotsapas, L. Devine, K. Raddassi, M. R. Ehlers, J. G. McNamara, K. M. Harris, S. Kanaparthi, D. Phippard, K. C. Herold, Changes in T-cell subsets identify responders to FcR-nonbinding anti-CD3 mAb (teplizumab) in patients with type 1 diabetes. Eur J Immunol 46, 230-241 (2016).
26. K. C. Herold, B. N. Bundy, S. A. Long, J. A. Bluestone, L. A. DiMeglio, M. J. Dufort, S. E. Gitelman, P. A. Gottlieb, J. P. Krischer, P. S. Linsley, J. B. Marks, W. Moore, A. Moran, H. Rodriguez, W. E. Russell, D. Schatz, J. S. Skyler, E. Tsalikian, D. K. Wherrett, A. G. Ziegler, C. J. Greenbaum, G. Type 1 Diabetes TrialNet Study, An Anti-CD3 Antibody, Teplizumab, in Relatives at Risk for Type 1 Diabetes. N Engl J Med 381, 603-613 (2019).
27. K. Nanto-Salonen, A. Kupila, S. Simell, H. Siljander, T. Salonsaari, A. Hekkala, S. Korhonen, R. Erkkola, J. I. Sipila, L. Haavisto, Nasal insulin to prevent type 1 diabetes in children with HLA genotypes and autoantibodies conferring increased risk of disease: a doubleblind, randomised controlled trial. The Lancet 372, 1746-1755 (2008).
28. G. Diabetes Prevention Trial-Type 1 Diabetes Study, Effects of insulin in relatives of patients with type 1 diabetes mellitus. N Engl J Med 346, 1685-1691 (2002).
29. G. Writing Committee for the Type 1 Diabetes TrialNet Oral Insulin Study, J. P. Krischer, D. A. Schatz, B. Bundy, J. S. Skyler, C. J. Greenbaum, Effect of Oral Insulin on Prevention of Diabetes in Relatives of Patients With Type 1 Diabetes: A Randomized Clinical Trial. JAMA 318, 1891-1902 (2017).
30. H. Elding Larsson, M. Lundgren, B. Jonsdottir, D. Cuthbertson, J. Krischer, A. -I. T. S. G. Di, Safety and efficacy of autoantigen-specific therapy with 2 doses of alum-formulated glutamate decarboxylase in children with multiple islet autoantibodies and risk for type 1 diabetes: A randomized clinical trial. Pediatr Diabetes 19, 410-419 (2018).
31. E. A. Gale, P. J. Bingley, C. L. Emmett, T. Collier, G. European Nicotinamide Diabetes Intervention Trial, European Nicotinamide Diabetes Intervention Trial (ENDIT): a randomised controlled trial of intervention before the onset of ty pe 1 diabetes. Lancet 363, 925-931 (2004).
32. D. L. Eizirik, M. L. Colli, F. Ortis, The role of inflammation in insulitis and beta-cell loss in type 1 diabetes. Nat Rev Endocrinol 5, 219-226 (2009).
33. E. B. Tsai, N. A. Sherry, J. P. Palmer, K. C. Herold, The rise and fall of insulin secretion in type 1 diabetes mellitus. Diabetologia 49, 261-270 (2006).
34. E. Ferrannini, A. Mari, V. Nofrate, J. M. Sosenko, J. S. Skyler, D. P. T. S. Group, Progression to diabetes in relatives of type 1 diabetic patients: mechanisms and mode of onset. Diabetes 59, 679-685 (2010).
35. K. C. Herold, S. Usmani-Brown, T. Ghazi, J. Lebastchi, C. A. Beam, M. D. Beilin, M. Ledizet, J. M. Sosenko, J. P. Krischer, J. P. Palmer, G. Type 1 Diabetes TrialNet Study, beta cell death and dysfunction during type 1 diabetes development in at-risk individuals. J Clin Invest 125, 1163-1173 (2015).
36. N. A. Sherry, E. B. Tsai, K. C. Herold, Natural history of beta-cell function in type 1 diabetes. Diabetes 54 Suppl 2, S32-39 (2005).
37. K. C. Herold, W. Hagopian, J. A. Auger, E. Poumian-Ruiz, L. Taylor, D. Donaldson, S. E. Gitelman, D. M. Harlan, D. Xu, R. A. Zivin, J. A. Bluestone, Anti-CD3 monoclonal antibody in new-onset type 1 diabetes mellitus. N Engl J Med 346, 1692-1698 (2002).
38. K. C. Herold, S. E. Gitelman, U. Masharani, W. Hagopian, B. Bisikirska, D. Donaldson, K. Rother, B. Diamond, D. M. Harlan, J. A. Bluestone, A Single Course of Anti- CD3 Monoclonal Antibody hOKT3 {gamma} 1 (Ala- Ala) Results in Improvement in C-Peptide Responses and Clinical Parameters for at Least 2 Years after Onset of Type 1 Diabetes. Diabetes 54, 1763-1769 (2005).
39. L. M. McLane, M. S. Abdel-Hakeem, E. J. Wherry, CD8 T Cell Exhaustion During Chronic Viral Infection and Cancer. Annu Rev Immunol 37, 457-495 (2019).
40. J. M. Sosenko, J. P. Palmer, C. J. Greenbaum, J. Mahon, C. Cowie, J. P. Krischer, H. P. Chase, N. H. White, B. Buckingham, K. C. Herold, D. Cuthbertson, J. S. Skyler, Patterns of metabolic progression to type 1 diabetes in the Diabetes Prevention Trial-Type 1. Diabetes Care 29, 643-649 (2006).
41. M. M. Bogun, B. N. Bundy, R. S. Goland, C. J. Greenbaum, C-Peptide Levels in Subjects Followed Longitudinally Before and After Type 1 Diabetes Diagnosis in TrialNet. Diabetes Care 43, 1-8 (2020).
42. L. Chatenoud, J. Primo, J. F. Bach, CD3 antibody-induced dominant self tolerance in overtly diabetic NOD mice. J Immunol 158, 2947-2954 (1997).
43. M. B. Davidson, A. L. Peters, D. L. Schriger, An alternative approach to the diagnosis of diabetes with a review of the literature. Diabetes Care 18, 1065-1071 (1995).
44. E. M. Akirav, M. T. Baquero, L. W. Opare-Addo, M. Akirav, E. Galvan, J. A. Kushner, D. L. Rimm, K. C. Herold, Glucose and inflammation control islet vascular density and betacell function in NOD mice: control of islet vasculature and vascular endothelial growth factor by glucose. Diabetes 60, 876-883 (2011).
45. N. A. Sherry, J. A. Kushner, M. Glandt, T. Kitamura, A. M. Brillantes, K. C. Herold, Effects of autoimmunity and immune therapy on beta-cell turnover in type 1 diabetes.
Diabetes 55, 3238-3245 (2006).
46. A. L. Perdigoto, P. Preston-Hurlburt, P. Clark, S. A. Long, P. S. Linsley, K. M. Harris, S. E. Gitelman, C. J. Greenbaum, P. A. Gottlieb, W. Hagopian, A. Woodwyk, J. Dziura, K. C. Herold, N. Immune Tolerance, Treatment of type 1 diabetes with teplizumab: clinical and immunological follow-up after 7 years from diagnosis. Diabetologia 62, 655-664 (2019).
47. K. C. Herold, S. L. Bucktrout, X. Wang, B. W. Bode, S. E. Gitelman, P. A. Gottlieb, J. Hughes, T. Joh, J. B. McGill, J. H. Pettus, S. Potluri, D. Schatz, M. Shannon, C. Udata, G. Wong, M. Levisetti, B. J. Ganguly, P. D. Garzone, R. N. W. Group, Immunomodulatory activity of humanized anti-IL-7R monoclonal antibody RN168 in subjects with type 1 diabetes. JCI Insight 4, (2019).
48. M. Battaglia, M. S. Anderson, J. H. Buckner, S. M. Geyer, P. A. Gottlieb, T. W. H. Kay, A. Lemmark, S. Muller, A. Pugliese, B. O. Roep, C. J. Greenbaum, M. Peakman, Understanding and preventing type 1 diabetes through the unique working model of TrialNet. Diabetologia 60, 2139-2147 (2017).
49. L. Yu, D. C. Boulware, C. A. Beam, J. C. Hutton, J. M. Wenzlau, C. J. Greenbaum, P. J. Bingley, J. P. Krischer, J. M. Sosenko, J. S. Skyler, G. S. Eisenbarth, J. L. Mahon, G. Type 1 Diabetes TrialNet Study, Zinc transporter-8 autoantibodies improve prediction of type 1 diabetes in relatives positive for the standard biochemical autoantibodies. Diabetes Care 35, 1213-1218 (2012).
50. A. American Diabetes, 2. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes 2019. Diabetes Care 42, S13-S28 (2019).
51. C. Steele, W. A. Hagopian, S. Gitelman, U. Masharani, M. Cavaghan, K. I. Rolher, D. Donaldson, D. M. Harlan, J. Bluestone, K. C. Herold, Insulin Secretion in Type 1 Diabetes. Diabetes 53, 426-433 (2004).
52. K. S. Polonsky, J. Licinio-Paixao, B. D. Given, W. Pugh, P Rue, J. Galloway, T. Karrison, B. Frank, Use of biosynthetic human C-peptide in the measurement of insulin secretion rates in normal volunteers and type I diabetic patients. J Clin Invest 77, 98-105 (1986).
53. E. Van Cauter, F. Mestrez, J. Sturis, K. S. Polonsky, Estimation of insulin secretion rates from C-peptide levels. Comparison of individual and standard kinetic parameters for C- peptide clearance. Diabetes 41, 368-377 (1992).
54. S. A. Long, J. Thorpe, K. C. Herold, M. Ehlers, S. Sanda, N. Lim, P. S. Linsley, G. T. Nepom, K. M. Harris, Remodeling T cell compartments during anti-CD3 immunotherapy of type 1 diabetes. Cell Immunol 319, 3-9 (2017).
55. Besser R.E.J., et al. “ISPAD clinical practice consensus guidelines 2022 : Stages of type 1 diabetes in children and adolescents” Pediatr. Diabetes 2022: 1-13
56. Classification and Diagnosis of Diabetes: Standard of Medical Care in Diabetes-2022. Diabetes Care 2022; 45 (Suppl. 1): S17-S38

Claims

CLAIMS What is claimed is:
1. A method of preventing or delaying onset of clinical type 1 diabetes (T1D), comprising: administering a prophylactically effective amount of an anti-CD3 antibody to a non-diabetic subject who is at risk of T1D, wherein the prophylactically effective amount has a cumulative dose of from about 10,500 pg/m2 to about 14,000 pg/ nr.
2. The method of claim 1, wherein the subject is 8 years of age or older.
3. The method of claim 1, wherein the subject is 5 years of age or older.
4. The method of claim 1, wherein the subject is 3 years of age or older.
5. The method of claim 1, wherein the subject is 1 year of age or older.
6. The method of claim 1, wherein the subject is 1 year old or younger.
7. The method of claim 1, wherein the non-diabetic subject is a relative of a patient with T1D.
8. The method of claim 1, further comprising determining that the non-diabetic subject (1) is negative for zinc transporter 8 (ZnT8) antibodies, (2) is HLA-DR4+, and/or (3) is not HLA-DR3+.
9. The method of claim 1, wherein the non-diabetic subject has 2 or more diabetes-related autoantibodies selected from islet cell antibodies (ICA), insulin autoantibodies (IAA), and antibodies to glutamic acid decarboxylase (GAD), tyrosine phosphatase (IA- 2/ICA512) or ZnT8.
10. The method of claim 1, wherein the non-diabetic subject has abnormal glucose tolerance on oral glucose tolerance test (OGTT).
11. The method of claim 10, wherein the abnormal glucose tolerance on OGTT is a fasting glucose level of 110-125 mg/dL, or 2 hour plasma of > 140 and < 200 mg/dL, or an intervening glucose value at 30, 60, or 90 minutes on OGTT > 200 mg/dL.
12. The method of claim 1, wherein the non-diabetic subject does not have antibodies against ZnT8.
13. The method of claim 1, wherein the non-diabetic subject is HLA-DR4+ and is not HLA- DR3+.
14. The method of claim 1, wherein the anti-CD3 antibody is selected from teplizumab, otelixizumab or foralumab. The method of claim 1, wherein the anti-CD3 antibody is teplizumab. The method of claim 14 or claim 15, wherein the prophylactically effective amount comprises a 14-day course of subcutaneous (SC) injection or intravenous (IV) infusion or oral administration of the anti-CD3 antibody with a cumulative dose of from about 11,000 pg/m2to about 14,000 pg/m2. The method of claim 14 or claim 15, comprising administering a 14-day course IV infusion at about 60 pg/m2 on day 1, about 125 pg/m2 on day 2, about 250 pg/m2 on day 3, and about 500 pg/m2 on day 4, and a dose of about 1,000 pg/m2 on each of days 5-14. The method of claim 14 or claim 15, comprising administering a 14-day course IV infusion at about 60 pg/m2, about 125 pg/m2 on day 1, about 250 pg/m2 on day 2, and about 500 pg/m2 on day 3, and a dose of about 1,030 pg/m2 on each of days 5-14. The method of claim 14 or claim 15, comprising administering a 14-day course IV infusion at about 100 pg/m2 on day 1, about 425 pg/m2 on day 2, about 850 pg/m2 on day 3, about 850 pg/m2 on day 4, and a dose of about 1,000 pg/m2 on each of days 5-14. The method of claim 14 or claim 15, comprising administering a 14-day course IV infusion at about 65 pg/m2 on day 1, about 125 pg/m2 on day 2, about 250 pg/m2 on day 3, and about 500 pg/m2 on day 4, and a dose of about 1,070 pg/m2 on each of days 5-14. The method of claim 14 or claim 15, comprising administering a 14-day course IV infusion at about 65 pg/m2 on day 1, about 125 pg/m2 on day 2, about 250 pg/m2 on day 3, and about 500 pg/m2 on day 4, and a dose of about 1,030 pg/m2 on each of days 5-14. The method of claim 14 or claim 15, wherein the prophylactically effective amount delays median time to clinical diagnosis of T1D by at least 50%, at least 80%, or at least 90%, or at least 12 months, at least 18 months, at least 24 months, at least 36 months, at least 48 months, or at least 60 months. The method of any one of claims 1-22, further comprising determining, prior to or after the administenng step, that the non-diabetic subject has more than about 10% TIGIT+KLRG1+CD8+ T-cells in all CD3+ T cells, which is indicative of successful prevention or delay of the onset of clinical T1D. The method of claim 23, wherein the determining of TIGIT+KLRG1+CD8+ T-cells is by flow cytometry. The method of any one of claims 1 -24, further comprising determining a decrease in a percentage of CD8+ T cells expressing proliferation markers Ki67 and/or CD57. A method of preventing or delaying onset of clinical type 1 diabetes (T1D), comprising: administering to a non-diabetic subject 8 years of age or older who is at risk of T1D a 14-day course IV infusion of teplizumab at about 65 pg/m2 on day 1, about 125 pg/m2 on day 2, about 250 pg/m2 on day 3, and about 500 pg/m2 on day 4, and a dose of about 1,030 pg/m2 on each of days 5-14. A method of delaying onset of Stage 3 type 1 diabetes (T1D), comprising: administering to a subject in need thereof who has a diagnosis of stage 2 T1D a 14-day course IV infusion of teplizumab at about 65 pg/m2 on day 1, about 125 pg/m2 on day 2, about 250 pg/m2 on day 3, and about 500 pg/m2 on day 4, and a dose of about 1,030 pg/m2 on each of days 5-14. The method of claim 27, wherein the subject in need thereof is an adult. The method of claim 27, wherein the subject in need thereof is a pediatric subject 8 years of age or older. The method of claim 27, wherein the subject in need thereof is a pediatric subject 5 years of age or older. The method of claim 27, wherein the subject in need thereof is a pediatric subject 3 years of age or older. The method of claim 27, wherein the subject in need thereof is a pediatric subject 1 year of age or older. The method of claim 27, wherein the subject in need thereof is a pediatric subject 1 year old or younger. The method of claim 27, the method comprising documenting two or more positive pancreatic islet autoantibodies in the subject who has dysglycemia without overt hyperglycemia before administering the 14-day course. The method of claim 27, wherein the subject in need thereof has dysglycemia without overt hyperglycemia and has two or more pancreatic islet autoantibodies. The method of claim 34 or claim 35, wherein the two or more pancreatic islet autoantibodies comprise islet cell antibodies (ICA), insulin autoantibodies (IAA), and antibodies to glutamic acid decarboxylase (GAD), tyrosine phosphatase (IA-2/ICA512) or ZnT8. The method of claim 27, the method comprising administering on at least each of days 1 -5, and prior to the administering of the 14-day course IV infusion, an effective amount of a nonsteroidal anti-inflammatory drug (NSAID), acetaminophen, an antihistamine, an antiemetic or a combination thereof. The method of claim 37, the method comprising administering orally the NSAID, acetaminophen, antihistamine, antiemetic or combination thereof. The method of claim 27, the method comprising administering the 14-day course IV infusion once daily for 14 consecutive days over a period of at least 30 minutes. A method of prognosing responsiveness of an anti-CD3 antibody in preventing or delaying onset of type 1 diabetes (T1D), the method comprising: administering a prophylactically effective amount of an anti-CD3 antibody to a non-diabetic subject who is at risk of T1D, wherein the prophylactically effective amount has a cumulative dose of about 10,500 pg/m2 to about 14,000 pg/m2; and determining C-peptide area under the curve (AUC): glucose AUC ratio, wherein an increase in the ratio indicates responsiveness to the anti-CD3 antibody and/or nonprogression to clinical T1D. An anti-CD3 antibody for use in a method of preventing or delaying onset of clinical type 1 diabetes (T1D), comprising: administering a prophylactically effective amount of an anti-CD3 antibody to a non-diabetic subject who is at risk of T1D, wherein the prophylactically effective amount has a cumulative dose of about 10,500 pg/m2 to about 14,000 pg/m2. A teplizumab composition for use in a method of delaying onset of Stage 3 type 1 diabetes (T1D), comprising: administering to a subject in need thereof who has a diagnosis of stage 2 T1D a 14-day course IV infusion at about 65 pg/m2 on day 1, about 125 pg/m2 on day 2, about 250 pg/m2 on day 3, and about 500 pg/m2 on day 4, and a dose of about 1,030 pg/m2 on each of days 5-14.
PCT/US2023/067357 2022-05-24 2023-05-23 Methods and compositions for preventing or delaying type 1 diabetes WO2023230476A1 (en)

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US202263345365P 2022-05-24 2022-05-24
US63/345,365 2022-05-24
US202263367992P 2022-07-08 2022-07-08
US63/367,992 2022-07-08
US202263382382P 2022-11-04 2022-11-04
US63/382,382 2022-11-04
US18/321,964 2023-05-23
TW112119143 2023-05-23
US18/321,964 US20230382993A1 (en) 2022-05-24 2023-05-23 Methods and compositions for preventing or delaying type 1 diabetes
TW112119143A TW202411250A (en) 2022-05-24 2023-05-23 Methods and compositions for preventing or delaying type 1 diabetes

Publications (1)

Publication Number Publication Date
WO2023230476A1 true WO2023230476A1 (en) 2023-11-30

Family

ID=88920022

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/067357 WO2023230476A1 (en) 2022-05-24 2023-05-23 Methods and compositions for preventing or delaying type 1 diabetes

Country Status (1)

Country Link
WO (1) WO2023230476A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140099318A1 (en) * 2011-05-21 2014-04-10 Macrogenics, Inc. CD3-Binding Molecules Capable of Binding to Human and Non-Human CD3
WO2021252917A2 (en) * 2020-06-11 2021-12-16 Provention Bio, Inc. Methods and compositions for preventing type 1 diabetes

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140099318A1 (en) * 2011-05-21 2014-04-10 Macrogenics, Inc. CD3-Binding Molecules Capable of Binding to Human and Non-Human CD3
WO2021252917A2 (en) * 2020-06-11 2021-12-16 Provention Bio, Inc. Methods and compositions for preventing type 1 diabetes

Similar Documents

Publication Publication Date Title
US11059896B2 (en) Stabilized formulations containing anti-interleukin-4 receptor (IL-4R) antibodies
US11434291B2 (en) Methods and compositions for preventing type 1 diabetes
US20220041720A1 (en) Methods and Compositions for Preventing Type 1 Diabetes
US20230382993A1 (en) Methods and compositions for preventing or delaying type 1 diabetes
WO2023230476A1 (en) Methods and compositions for preventing or delaying type 1 diabetes
TW202411250A (en) Methods and compositions for preventing or delaying type 1 diabetes
US20210393779A1 (en) Activin a antibody formulations and methods of use thereof
US20220372146A1 (en) Methods for treating post infectious autoimmune diabetes
US20220380465A1 (en) Methods for treating type 1 diabetes
EP4164689A2 (en) Methods and compositions for preventing type 1 diabetes
EP3873601A1 (en) Methods and compositions for preventing type 1 diabetes
AU2022347193A1 (en) Methods for prognosing type 1 diabetes treatments
WO2023039295A1 (en) Methods and compositions comprising anti-cd3 antibodies and dyrk1a inhibitors for treating diabetes

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23812720

Country of ref document: EP

Kind code of ref document: A1