AU2002249768A1 - Method of determining a chemotherapeutic regimen based on ERCC1 expression - Google Patents

Method of determining a chemotherapeutic regimen based on ERCC1 expression

Info

Publication number
AU2002249768A1
AU2002249768A1 AU2002249768A AU2002249768A AU2002249768A1 AU 2002249768 A1 AU2002249768 A1 AU 2002249768A1 AU 2002249768 A AU2002249768 A AU 2002249768A AU 2002249768 A AU2002249768 A AU 2002249768A AU 2002249768 A1 AU2002249768 A1 AU 2002249768A1
Authority
AU
Australia
Prior art keywords
erccl
mrna
sample
tumor
gene
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU2002249768A
Other versions
AU2002249768B2 (en
Inventor
Kathleen D. Danenberg
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Response Genetics Inc
Original Assignee
Response Genetics 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
Priority claimed from US09/796,491 external-priority patent/US6518416B1/en
Priority claimed from US09/988,784 external-priority patent/US6602670B2/en
Application filed by Response Genetics Inc filed Critical Response Genetics Inc
Publication of AU2002249768A1 publication Critical patent/AU2002249768A1/en
Application granted granted Critical
Publication of AU2002249768B2 publication Critical patent/AU2002249768B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Description

METHOD OF DETERMINING A CHEMOTHERAPEUTIC
REGIMEN BASED ON ERCCl EXPRESSION
FIELD OF THE INVENTION
The present invention relates to prognostic methods which are useful in
medicine, particularly cancer chemotherapy. More particularly, the invention
relates to assessment of tumor cell gene expression in a patient. The survival of
patients treated with chemotherapeutic agents that target DNA, especially agents that
damage DNA in the manner of platinating agents is assayed by examining the mRNA expressed from genes involved in DNA repair in humans.
BACKGROUND OF THE INVENTION
Cancer arises when a normal cell undergoes neoplastic transformation and
becomes a malignant cell. Transformed (malignant) cells escape normal physiologic
controls specifying cell phenotype and restraining cell proliferation. Transformed
cells in an individual's body thus proliferate, forming a tumor. When a tumor is
found, the clinical objective is to destroy malignant cells selectively while mitigating
any harm caused to normal cells in the individual undergoing treatment.
Chemotherapy is based on the use of drugs that are selectively toxic
(cytotoxic) to cancer cells. Several general classes of chemotherapeutic drugs have
been developed, including drugs that interfere with nucleic acid synthesis, protein
synthesis, and other vital metabolic processes. These generally are referred to as
antimetabolite drugs. Other classes of chemotherapeutic drugs inflict damage on
cellular DNA. Drugs of these classes generally are referred to as geno toxic. Susceptibility of an individual neoplasm to a desired chemotherapeutic drug or
combination of drugs often, however, can be accurately assessed only after a trial
period of treatment. The time invested in an unsuccessful trial period poses a
significant risk in the clinical management of aggressive malignancies.
The repair of damage to cellular DNA is an important biological process
carried out by a cell's enzymatic DNA repair machinery. Unrepaired lesions in a
cell's genome can impede DNA replication, impair the replication fidelity of newly
synthesized DNA and/or hinder the expression of genes needed for cell survival.
Thus, genotoxic drugs generally are considered more toxic to actively dividing cells
that engage in DNA synthesis than to quiescent, nondividing cells. Normal cells of
many body tissues are quiescent and commit infrequently to re-enter the cell cycle
and divide. Greater time between rounds of cell division generally is afforded for the
repair of DNA damage in normal cells inflicted by chemotherapeutic genotoxins. As a result, some selectivity is achieved for the killing of cancer cells. Many treatment
regimens reflect attempts to improve selectivity for cancer cells by co administering chemotherapeutic drugs belonging to two or more of these general classes.
Because effective chemotherapy in solid tumors usually requires a
combination of agents, the identification and quantification of determinants of
resistance or sensitivity to each single drug has become an important tool to design
individual combination chemotherapy.
Two widely used genotoxic anticancer drugs that have been shown to
damage cellular DNA are cisplatin (DDP) and carboplatin. Cisplatin and/or
carboplatin currently are used in the treatment of selected, diverse neoplasms of
epithelial and mesenchymal origin, including carcinomas and sarcomas of the
respiratory, gastrointestinal and reproductive tracts, of the central nervous system, and of squamous origin in the head and neck. Cisplatin in combination with other
agents is currently preferred for the management of testicular carcinoma, and in
many instances produces a lasting remission. (Loehrer etal., 1984, 100 Ann. Int.
Med. 704). Cisplatin (DDP) disrupts DNA structure through formation of
intrastrand adducts. Resistance to platinum agents such as DDP has been attributed
to enhanced tolerance to platinum adducts, decreased drug accumulation, or
enhanced DNA repair. Although resistance to DDP is multifactoral, alterations in
DNA repair mechanisms probably play a significant role. Excision repair of bulky
DNA adducts, such as those formed by platinum agents, appears to be mediated by
genes involved in DNA damage recognition and excision. Cleaver et al.,
Carcinogenesis 11:875-882 (1990); Hoeijmakers et al., Cancer Cells 2:311-320
(1990); Shivji et al., Cell 69:367-374 (1992). Indeed, cells carrying a genetic defect
in one or more elements of the enzymatic DNA repair machinery are extremely sensitive to cisplatin. Fraval et al. (1978), 51 Mutat. Res. 121, Beck and Brubaker
(1973), 116 J. Bacteriol 1247.
The excision repair cross-complementing (ERCCl) gene is essential in the
repair of DNA adducts. The human ERCCl gene has been cloned. Westerveld et al.,
Nature (London) 310:425-428 (1984); Tanaka et al., Nature 348:73-76 (1990).
Several studies using mutant human and hamster cell lines that are defective in this
gene and studies in human tumor tissues indicate that the product encoded by
ERCCl is involved in the excision repair of platinum-DNA adducts. Dabholkar et
al., J. Natl. Cancer Inst. 84:1512-1517 (1992); Dijt et al, Cancer Res. 48:6058-6062
(1988); Hansson et al, Nucleic Acids Res. 18: 35-40 (1990).
When transfected into DNA-repair deficient CHO cells, ERCCl confers
cellular resistance to cisplatin along with the ability to repair platinum-DNA adducts. Hansson et al., Nucleic Acids Res. 18: 35-40 (1990). Currently accepted
models of excision repair suggest that the damage recognition/excision step is rate-
limiting to the excision repair process.
The relative levels of expression of excision repair genes such as ERCCl in
malignant cells from cancer patients receiving platinum-based therapy has been
examined. Dabholkar et al., J. Natl. Cancer Inst. 84:1512-1517 (1992). ERCCl
overexpression in gastric cancer patients has been reported to have a negative impact
on tumor response and ultimate survival when treated with the chemotherapeutic
regimen of cisplatin (DDP)/fluorouracil (Metzger, et l., J Gin Oncol 16: 309,
1998). Recent evidence indicates that gemcitabine (Gem) may modulate ERCCl
nucleotide excision repair (NER) activity. Thus, intratumoral levels of ERCCl expression may be a major prognostic factor for determining whether or not DDP
and GEM would be an effective therapeutic cancer patients.
Most pathological samples are routinely fixed and paraffin-embedded (FPE)
to allow for histo logical analysis and subsequent archival storage. Thus, most
biopsy tissue samples are not useful for analysis of gene expression because such
studies require a high integrity of RNA so that an accurate measure of gene
expression can be made. Currently, gene expression levels can be only qualitatively
monitored in such fixed and embedded samples by using immunohistochemical
staining to monitor protein expression levels.
Until now, quantitative gene expression studies including those of ERCCl
expression have been limited to reverse transcriptase polymerase chain reaction
(RT-PCR) amplification of RNA from fresh or frozen tissue. U.S. Patent No.
5,705,336 to Reed et al., discloses a method of quantifying ERCCl mRNA from
ovarian tumor tissue and determining whether that tissue will be sensitive to platinum-based chemotherapy. Reed et al., quantify ERCCl mRNA from frozen
ovarian tumor biopsies.
The use of frozen tissue by health care professionals as described in Reed et
al., poses substantial inconveniences. Rapid biopsy delivery to avoid tissue and
subsequent mRNA degradation is the primary concern when planning any RNA-
based quantitative genetic marker assay. The health care professional performing
the biopsy, must hastily deliver the tissue sample to a facility equipped to perform an
RNA extraction protocol immediately upon tissue sample receipt. If no such facility
is available, the clinician must promptly freeze the sample in order to prevent
mRNA degradation. In order for the diagnostic facility to perform a useful RNA
extraction protocol prior to tissue and RNA degradation, the tissue sample must
remain frozen until it reaches the diagnostic facility, however far away that may be. Maintenance of frozen tissue integrity during transport using specialized couriers
equipped with liquid nitrogen and dry ice, comes only at a great expense.
Routine biopsies generally comprise a heterogenous mix of stromal and
tumorous tissue. Unlike with fresh or frozen tissue, FPE biopsy tissue samples are
readily microdissected and separated into stromal and tumor tissue and therefore,
offer andvantage over the use of fresh or frozen tissue. However, isolation of RNA
from fixed tissue, and especially fixed and paraffin embedded tissue, results in
highly degraded RNA, which is generally not applicable to gene expression studies.
A number of techniques exist for the purification of RNA from biological
samples, but none is reliable for isolation of RNA from FPE samples. For example,
Chomczynski (U.S. Pat. No. 5,346,994) describes a method for purifying RNA from
tissues based on a liquid phase separation using phenol and guanidine
isothiocyanate. A biological sample is homogenized in an aqueous solution of phenol and guanidine isothiocyanate and the homogenate thereafter mixed with
chloroform. Following centrifugation, the homogenate separates into an organic
phase, an interphase and an aqueous phase. Proteins are sequestered in the organic
phase, DNA in the interphase, and RNA in the aqueous phase. RNA can be
precipitated from the aqueous phase. Unfortunately, this method is not applicable to
fixed and paraffin-embedded (FPE) tissue samples.
Other known techniques for isolating RNA typically utilize either guanidine
salts or phenol extraction, as described for example in Sambrook, J. et al, (1989) at
pp. 7.3-7.24, and in Ausubel, F. M. et al, (1994) at pp. 4.0.3-4.4.7. Again, none of
the known methods provides reproducible quantitative results in the isolation of
RNA from paraffin-embedded tissue samples.
Techniques for the isolation of RNA from paraffin-embedded tissues are thus
particularly needed for the study of gene expression in tumor tissues, since expression levels of certain receptors or enzymes can be used to determine the
likelihood of success of a particular treatment.
There is a need for a method of quantifying ERCCl mRNA from
paraffinized tissue in order to provide an early prognosis for proposed genotoxic
cancer therapies. As a result, there has been a concerted yet unsuccessful effort in
the art to obtain a quantification of ERCCl expression in fixed and paraffinized
(FPE) tissue. Accordingly, it is the object of the invention to provide a method for
assessing ERCCl levels in tissues fixed and paraffin-embedded (FPE) and
prognosticate the probable resistance of a patient's tumor to treatment with DNA
damaging agents, creating the type of lesions in DNA that are created by DNA
platinating agents, by examination of the amount of ERCCl mRNA in a patient's
tumor cells and comparing it to a predetermined threshold expression level. SUMMARY OF THE INVENTION
In one aspect of the invention there is provided a method for assessing levels
of expression of ERCCl mRNA obtained from fixed and paraffin-embedded (FPE)
fixed and paraffin-embedded (FPE) tumor cells.
In another aspect of the invention there is provided a method of quantifying
the amount of ERCCl mRNA expression relative to an internal control from a fixed
and paraffin-embedded (FPE) tissue sample. This method includes isolation of total
mRNA from said sample and determining the quantity of ERCCl mRNA relative to
the quantity of an internal control gene's mRNA.
In- an embodiment of this aspect of the invention, there are provided
oligonucleotide primers having the sequence of ERCC1-504F (SEQ ID NO: 1) or
ERCC1-574R (SEQ ID NO:2) and sequences substantially identical thereto. The
invention also provides for oligonucleotide primers having a sequence that
hybridizes to SEQ ID NO: 1 or SEQ ID NO:2 or their complements under stringent
conditions.
In yet another aspect of the invention there is provided a method for
determining a chemotherapeutic regimen for a patient, comprising isolating RNA
from a fixed and paraffin-embedded (FPE) tumor sample; determining a gene
expression level of ERCCl in the sample; comparing the ERCCl gene expression
levels in the sample with a predeterimined threshold level for the ERCCl gene; and
determining a chemotherapeutic regimen based on results of the comparison of the
ERCCl gene expression level with the predetermined threshold level.
The invention further relates to a method of normalizing the uncorrected
gene expression (UGE) of ERCCl relative to an internal control gene in a tissue sample analyzed using TaqMan® technology to known ERCCl expression levels
relative to an internal control from samples analyzed by pre-TaqMan® technology.
DESCRIPTION OF THE DRAWING
Figure 1 is a graph showing the overall survival of patients receiving
Cisplatin/Gem treatment vs. Corrected Relative ERCCl Expression in NSCLC.
Patient Corrected Relative ERCCl Expression levels lower than the threshold of 6.7
x 10"3 correlated with significantly better survival. While patient Corrected Relative
ERCCl Expression levels higher than the threshold of 6.7 x 10"3 correlated with
significantly worse survival. (P=0.009 Log rank test)
Figure 2 is a chart illustrating how to calculate Corrected Relative ERCCl
expression relative to an internal control gene. The chart contains data obtained with
two test samples, (unknowns 1 and 2), and illustrates how to determine the
uncorrected gene expression data (UGE). The chart also illustrates how to
normalize UGE generated by the TaqMan® instrument with known relative ERCCl
values determined by pre-TaqMan® technology. This is accomplished by
multiplying UGE to a correction factor KERCCJ. The internal control gene in the
figure is β-actin and the calibrator RNA is Human Liver Total RNA (Stratagene,
Cat. #735017).
Figure 3 is a table showing the demographic details of the 56 patients in the
study, tumor stage and cell types. The median number of treatment cycles received
was 3 (range 1-6). Fourteen patients (25%) had previously received chemotherapy,
mostly (9 patients) taxane therapy alone or in combination with DDP or carboplatin.
Three of the 56 patients had received radiotherapy and 5 patients had under gone
surgical resection of the primary tumor. Figure 4 is a table showing patients with Corrected ERCCl expression levels
below the threshold had a significantly longer median survival of 61.6 weeks (95%
CI. 42.4, 80.7 weeks) compared to 20.4 weeks (95% CI. 6.9, 33.9 weeks) for
patients with Corrected ERCCl levels above the threshold. Adjusted for tumor stage,
the log rank statistic for the association between low or high ERCCl expression and
overall survival was 3.97 and the P value was 0.046. The unadjusted log rank results
are shown in this figure. Also shown are factors that were significantly associated
with overall survival on univariable analysis using Kaplan Meier survival curves, and
the log rank test. These were the presence of pretreatment weight loss and the
ECOG performance status. Patient age (P=0.18), sex (P=0.87), tumor stage
(P=0.99), tumor cell type (P=0.63), and presence of pleural effusion (P=0.71) were
not significant prognostic factors for overall survival. Corrected Relative ERCCl Expression level, ECOG performance status, and weight loss remained significant
prognostic factors for survival in the Cox proportional hazards regression model
multivariable analysis. P values for a Cox regression model stratified on tumor stage
were 0.038 for ERCCl, 0.017 for weight loss, and 0.02 for ECOG performance
status (PS 0 versus 1 or 2).
DETAILED DESCRIPTION OF THE INVENTION
The present invention resides in part in the finding that the amount of
ERCCl mRNA in a tumor correlates with survival in patients treated with DNA
platinating agents. Patients with tumors expressing high levels of ERCCl mRNA are
considered likely to be resistant to platinum-based chemotherapy and this have lower
levels of survivability. Conversely, those patients whose tumors expressing low
amounts of ERCCl mRNA are likely to be sensitive to platinum-based
chemotherapy and have greater levels of survivability. A patient's relative
expression of tumor ERCCl mRNA is judged by comparing it to a predetermined
threshold expression level.
The invention relates to a method of quantifying the amount of ERCCl
mRNA expression in fixed and paraffin-embedded (FPE) tissue relative to gene
expression of an internal control. The present inventors have developed
oligonucleotide primers that allow accurate assessment of ERCCl expression in
tissues that have been fixed and embedded. The invention oligonucleotide primers,
ERCC1-504F (SEQ ID NO: 1), ERCC1-574R (SEQ ID NO: 2), or oligonucleotide
primers substantially identical thereto, preferably are used together with RNA
extracted from fixed and paraffin embedded (FPE) tumor samples. This
measurement of ERCCl gene expression may then be used for prognosis of
platinum-based chemotherapy.
This embodiment of the invention involves first, a method for reliable
extraction of RNA from an FPE sample and second, determination of the content of
ERCCl mRNA in the sample by using a pair of oligonucleotide primers, preferably
oligionucleotide primer pair ERCC1-504F (SEQ ID NO: 1) and ERCC1-574R (SEQ ID NO: 2), or oligonucleotides substantially identical thereto, for carrying out
reverse transcriptase polymerase chain reaction. RNA is extracted from the FPE
cells by any of the methods for mRNA isolation from such samples as described in
US Patent Application No. 09/469,338, filed December 20, 1999, and is hereby
incorporated by reference in its entirety.
The present method can be applied to any type of tissue from a patient. For
examination of resistance of tumor tissue, it is preferable to examine the tumor
tissue. In a preferred embodiment, a portion of normal tissue from the patient from
which the tumor is obtained, is also examined. Patients whose normal tissues are
expected to be resistant to platinum-based chemotherapeutic compounds, i.e., show
a high level of ERCCl gene expression, but whose tumors are expected to be
sensitive to such compounds, i.e., show a low level of ERCCl gene expression, may then be treated with higher amounts of the chemotherapeutic composition.
Patients showing a level of ERCCl gene expression below the threshold
level, may be treated with higher amounts of the chemotherapeutic composition
because they are expected to have greater survivability than patients with tumors
expressing a level of ERCCl gene expression above the threshold level.
Alternatively, the clinician may determine that patients with tumors expressing a
level of ERCCl gene expression above the threshold level may not derive any
significant benefit from chemotherapy given their low expected survivability.
The methods of the present invention can be applied over a wide range of
tumor types. This allows for the preparation of individual "tumor expression
profiles" whereby expression levels of ERCCl are determined in individual patient
samples and response to various chemotherapeutics is predicted. Preferably, the methods of the invention are applied to solid tumors, most preferably Non-Small
Cell Lung Cancer (NSCLC) tumors. For application of some embodiments of the
invention to particular tumor types, it is preferable to confirm the relationship of
ERCCl gene expression levels to survivability by compiling a dataset that enables
correlation of a particular ERCCl expression and clinical resistance to platinum-
based chemotherapy.
A "predetermined threshold level", as defined herein, is a corrected relative
level of ERCCl tumor expression above which it has been found that tumors are
likely to be resistant to a platinum-based chemotherapeutic regimen. Tumor
expression levels below this threshold level are likely to be found in tumors sensitive
to platinum-based chemotherapeutic regimen. The range of corrected relative
expression of ERCCl, expressed as a ratio of ERCCl : β-actin, among tumors
responding to a platinum-based chemotherapeutic regimen is less than about 6.7 x
10"3. Tumors that do not respond to a platinum-based chemotherapeutic regimen
have relative expression of ERCCl : β-actin ratio above about 6.7 x 10"3. See
Example 4.
A "predetermined threshold level" is further defined as tumor corrected
relative ERCCl expression levels above which patients receiving a platinum-based
chemotherapeutic regimen are likely to have low survivability. Tumor corrected
relative ERCCl expression levels below this threshold level in patients receiving a
platinum-based chemotherapeutic regimen correlate to high patient survivability.
The threshold corrected relative ERCCl expression, expressed as a ratio of ERCCl :
β-actin, is about 6.7 x 10"3. Figure 1, see Example 4. However, the present invention
is not limited to the use of β-actin as an internal control gene. In performing the method of this embodiment of the present invention, tumor
cells are preferably isolated from the patient. Solid or lymphoid tumors or portions
thereof are surgically resected from the patient or obtained by routine biopsy. RNA
isolated from frozen or fresh samples is extracted from the cells by any of the
methods typical in the art, for example, Sambrook, Fischer and Maniatis, Molecular
Cloning, a laboratory manual, (2nd ed.), Cold Spring Harbor Laboratory Press, New
York, (1989). Preferably, care is taken to avoid degradation of the RNA during the
extraction process.
However, tissue obtained from the patient after biopsy is often fixed, usually
by formalin (formaldehyde) or gluteraldehyde, for example, or by alcohol
immersion. Fixed biological samples are often dehydrated and embedded in paraffin
or other solid supports known to those of skill in the art. Non-embedded, fixed tissue
may also be used in the present methods. Such solid supports are envisioned to be removable with organic solvents for example, allowing for subsequent rehydration
of preserved tissue.
RNA is extracted from the FPE cells by any of the methods as described in
US Patent Application No. 09/469,338, filed December 20, 1999, which is hereby
incorporated by reference in its entirety. Fixed and paraffin-embedded (FPE) tissue
samples as described herein refers to storable or archival tissue samples. RNA may
be isolated from an archival pathological sample or biopsy sample which is first
deparaffinized. An exemplary deparaffinization method involves washing the
paraffinized sample with an organic solvent, such as xylene, for example.
Deparaffinized samples can be rehydrated with an aqueous solution of a lower
alcohol. Suitable lower alcohols, for example include, methanol, ethanol, propanols, and butanols. Deparaffinized samples may be rehydrated with successive washes
with lower alcoholic solutions of decreasing concentration, for example.
Alternatively, the sample is simultaneously deparaffinized and rehydrated. RNA is
then extracted from the sample.
For RNA extraction, the fixed or fixed and deparaffinized samples can be
homogenized using mechanical, sonic or other means of homogenization.
Rehydrated samples may be homogenized in a solution comprising a chaotropic
agent, such as guanidinium thiocyanate (also sold as guanidinium isothiocyanate).
Homogenized samples are heated to a temperature in the range of about 50 to about
100 °C in a chaotropic solution, which contains an effective amount of a chaotropic
agent, such as a guanidinium compound. A preferred chaotropic agent is
guanidinium thiocyanate.
An "effective concentration of chaotropic agent" is chosen such that at an
RNA is purified from a paraffin-embedded sample in an amount of greater than
about 10-fold that isolated in the absence of a chaotropic agent. Chaotropic agents
include: guanidinium compounds, urea, formamide, potassium iodiode, potassium
thiocyantate and similar compounds. The preferred chaotropic agent for the methods
of the invention is a guanidinium compound, such as guanidinium isothiocyanate
(also sold as guanidinium thiocyanate) and guanidinium hydro chloride. Many
anionic counterions are useful, and one of skill in the art can prepare many
guanidinium salts with such appropriate anions. The effective concentration of
guanidinium solution used in the invention generally has a concentration in the range
of about 1 to about 5M with a preferred value of about 4M. If RNA is already in
solution, the guanidinium solution may be of higher concentration such that the final concentration achieved in the sample is in the range of about 1 to about 5M. The
guanidinium solution also is preferably buffered to a pH of about 3 to about 6, more
preferably about 4, with a suitable biochemical buffer such as Tris-Cl. The
chaotropic solution may also contain reducing agents, such as dithiothreitol (DTT)
and β-mercaptoethanol (BME). The chaotropic solution may also contain RNAse
inhibitors.
Homogenized samples may be heated to a temperature in the range of about
50 to about 100 °C in a chaotropic solution, which contains an effective amount of a
chaotropic agent, such as a guanidinium compound. A preferred chaotropic agent is
guanidinium thiocyanate.
RNA is then recovered from the solution by, for example, phenol chloroform
extraction, ion exchange chromatography or size-exclusion chromatography. RNA
may then be further purified using the techniques of extraction, electrophoresis,
chromatography, precipitation or other suitable techniques.
The quantification of ERCCl mRNA from purified total mRNA from fresh,
frozen or fixed is preferably carried out using reverse-transcriptase polymerase chain
reaction (RT-PCR) methods common in the art, for example. Other methods of
quantifying of ERCCl mRNA include for example, the use of molecular beacons
and other labeled probes useful in multiplex PCR. Additionally, the present
invention envisages the quantification of ERCCl mRNA via use of PCR-free
systems employing, for example fluorescent labeled probes similar to those of the
Invader® Assay (Third Wave Technologies, Inc.). Most preferably, quantification of
ERCCl cDNA and an internal control or house keeping gene (e.g. β-actin) is done
using a fluorescence based real-time detection method (ABI PRISM 7700 or 7900 Sequence Detection System [TaqMan®], Applied Biosystems, Foster City, CA.) or
similar system as described by Heid et al, (Genome Res 1996;6:986-994) and
Gibson et ^ .(Genome Res 1996;6:995-1001). The output of the ABI 7700
(TaqMan® Instrument) is expressed in Ct's or "cycle thresholds". With the
TaqMan® system, a highly expressed gene having a higher number of target
molecules in a sample generates a signal with fewer PCR cycles (lower Ct) than a
gene of lower relative expression with fewer target molecules (higher Ct).
As used herein, a "house keeping" gene or "internal control" is meant to
include any constitutively or globally expressed gene whose presence enables an
assessment of ERCCl mRNA levels. Such an assessment comprises a determination
of the overall constitutive level of gene transcription and a control for variations in
RNA recovery. "House-keeping" genes or "internal controls" can include, but are
not limited to the cyclophilin gene, β-actin gene, the transferrin receptor gene,
GAPDH gene, and the like. Most preferably, the internal control gene is β-actin
gene as described by Eads et al, Cancer Research 1999; 59:2302-2306.
A control for variations in RNA recovery requires the use of "calibrator
RNA." The "calibrator RNA" is intended to be any available source of accurately
pre-quantified control RNA. Preferably, Human Liver Total RNA (Stratagene, Cat.
#735017) is used.
"Uncorrected Gene Expression (UGE)" as used herein refers to the numeric
output of ERCCl expression relative to an internal control gene generated by the
TaqMan® instrument. The equation used to determine UGE is shown in Example 3,
and illustrated with sample calculations in Figure 2.
A further aspect of this invention provides a method to normalize uncorrected gene expression (UGE) values acquired from the TaqMan® instrument
with "known relative gene expression" values derived from non-TaqMan®
technology. Preferably, the known non-TaqMan® derived relative ERCCl : β-
actin expression values are normalized with TaqMan® derived ERCCl UGE values
from a tissue sample.
"Corrected Relative ERCCl Expression" as used herein refers to normalized
ERCCl expression whereby UGE is multiplied with a ERCCl specific correction
factor (KERC ), resulting in a value that can be compared to a known range of
ERCCl expression levels relative to an internal control gene. Example 3 and Figure
2 illustrate these calculations in detail. These numerical values allow the
determination of whether or not the "Corrected Relative ERCCl Expression" of a
particular sample falls above or below the "predetermined threshold" level. The
predetermined threshold level of Corrected Relative ERCCl Expression to β-actin
level is about 6.7 x 10"3. ¥L.mccl specific for ERCCl, the internal control β-actin and
calibrator Human Liver Total RNA (Stratagene, Cat. #735017), is 1.54 x 10'3.
"Known relative gene expression" values are derived from previously
analyzed tissue samples and are based on the ratio of the RT-PCR signal of a target
gene to a constitutively expressed internal control gene (e.g. β-Actin, GAPDH, etc.).
Preferably such tissue samples are formalin fixed and paraffin-embedded (FPE)
samples and RNA is extracted from them according to the protocol described in
Example 1 and in US Patent Application No. 09/469,338, filed December 20, 1999,
which is hereby incorporated by reference in its entirety. To quantify gene
expression relative to an internal control standard quantitative RT-PCR technology
known in the art is used. Pre-TaqMan® technology PCR reactions are run for a fixed number of cycles (i.e., 30) and endpoint values are reported for each sample.
These values are then reported as a ratio of ERCCl expression to β-actin expression.
See U.S. Patent No. 5,705,336 to Reed et al.
KgRca may be determined for an internal control gene other than β-actin
and/or a calibrator RNA different than Human Liver Total RNA (Stratagene, Cat.
#735017). To do so, one must calibrate both the internal control gene and the
calibrator RNA to tissue samples for which ERCCl expression levels relative to that
particular internal control gene have already been determined (i.e., "known relative
gene expression"). Preferably such tissue samples are formalin fixed and paraffin-
embedded (FPE) samples and RNA is extracted from them according to the protocol
described in Example 1 and in US Patent Application No. 09/469,338, filed
December 20, 1999, which is hereby incorporated by reference in its entirety. Such
a determination can be made using standard pre-TaqMan®, quantitative RT-PCR
techniques well known in the art. Upon such a determination, such samples have
"known relative gene expression" levels of ERCCl useful in the determining a new
KmccJ specific for the new internal control and/or calibrator RNA as described in
Example 3.
The methods of the invention are applicable to a wide range of tissue and
tumor types and so can be used for assessment of clinical treatment of a patient and
as a diagnostic or prognostic tool for a range of cancers including breast, head and
neck, lung, esophageal, colorectal, and others. In a preferred embodiment, the
present methods are applied to prognosis of Non-Small Cell Lung Cancer (NSCLC).
Pre-chemotherapy treatment tumor biopsies are usually available only as
fixed paraffin embedded (FPE) tissues, generally containing only a very small amount of heterogeneous tissue. Such FPE samples are readily amenable to
microdissection, so that ERCCl gene expression may be determined in tumor tissue
uncontaminated with stromal tissue. Additionally, comparisons can be made
between stromal and tumor tissue within a biopsy tissue sample, since such samples
often contain both types of tissues.
Generally, any oligonucleotide pair that flanks a region of ERCCl gene may
be used to carry out the methods of the invention. Primers hybridizing under
stringent conditions to a region of the ERCCl gene for use in the present invention
will amplify a product between 20-1000 base pairs, preferably 50-100 base pairs,
most preferably less than 100 base pairs.
The invention provides specific oligonucleotide primers pairs and
oligonucleotide primers substantially identical thereto, that allow particularly
accurate assessment of ERCCl expression in FPE tissues. Preferable are
oligonucleotide primers, ERCC1-504F (SEQ ID NO: 1) and ERCCl (SEQ ID NO:
2), (also referred to herein as the oligonucleotide primer pair ERCCl) and
oligonucleotide primers substantially identical thereto. The oliogonucleotide
primers ERCC1-504F (SEQ ID NO: 1) and ERCCl, (SEQ ID NO: 2) hybridize to
the ERCCl gene (SEQ ID NO: 7) under stringent conditions and have been shown to
be particularly effective for measuring ERCCl mRNA levels using RNA extracted
from the FPE cells by any of the methods for mRNA isolation, for example as
described Example 1 and in US Patent Application No. 09/469,338, filed December
20, 1999, which is hereby incorporated by reference in its entirety.
"Substantially identical" in the nucleic acid context as used herein, means
hybridization to a target under stringent conditions, and also that the nucleic acid segments, or their complementary strands, when compared, are the same when
properly aligned, with the appropriate nucleotide insertions and deletions, in at least
about 60% of the nucleotides, typically, at least about 70%, more typically, at least
about 80%, usually, at least about 90%, and more usually, at least, about 95-98% of
the nucleotides. Selective hybridization exists when the hybridization is more
selective than total lack of specificity. See, Kanehisa, Nucleic Acids Res., 12:203-
213 (1984).
This invention includes substantially identical oligonucleotides that
hybridize under stringent conditions (as defined herein) to all or a portion of the
oligonucleotide primer sequence of ERCC1-504F (SEQ ID NO: 1), its complement
or ERCC1-574R (SEQ ID NO: 2), or its complement.
Under stringent hybridization conditions, only highly complementary, i.e.,
substantially similar nucleic acid sequences hybridize. Preferably, such conditions prevent hybridization of nucleic acids having 4 or more mismatches out of 20
contiguous nucleotides, more preferably 2 or more mismatches out of 20 contiguous
nucleotides, most preferably one or more mismatch out of 20 contiguous
nucleotides.
The hybridizing portion of the nucleic acids is typically at least 10 (e.g., 15)
nucleotides in length. The hybridizing portion of the hybridizing nucleic acid is at
least about 80%, preferably at least about 95%, or most preferably about at least
98%, identical to the sequence of a portion or all of oligonucleotide primer ERCCl -
504F (SEQ ID NO: 1), its complement or ERCC1-574R (SEQ ID NO: 2), or its
complement.
Hybridization of the oligonucleotide primer to a nucleic acid sample under stringent conditions is defined below. Nucleic acid duplex or hybrid stability is
expressed as a melting temperature (T^, which is the temperature at which the probe
dissociates from the target DNA. This melting temperature is used to define the
required stringency conditions. If sequences are to be identified that are
substantially identical to the probe, rather than identical, then it is useful to first
establish the lowest temperature at which only homologous hybridization occurs
with a particular concentration of salt (e.g. SSC or SSPE). Then assuming that 1%
mismatching results in a 1°C decrease in Tm, the temperature of the final wash in the
hybridization reaction is reduced accordingly (for example, if sequences having
>95% identity with the probe are sought, the final wash temperature is decrease by
5° C). In practice, the change in Tm can be between 0.5°C and 1.5°C per 1% mismatch.
Stringent conditions involve hybridizing at 68° C in 5x SSC/5x Denhart's
solution/1.0% SDS, and washing in 0.2x SSC/0.1% SDS at room temperature.
Moderately stringent conditions include washing in 3x SSC at 42° C The
parameters of salt concentration and temperature be varied to achieve optimal level
of identity between the primer and the target nucleic acid. Additional guidance
regarding such conditions is readily available in the art, for example, Sambrook,
Fischer and Maniatis, Molecular Cloning, a laboratory manual, (2nd ed.), Cold
Spring Harbor Laboratory Press, New York, (1989) and F. M. Ausubel et al eds.,
Current Protocols in Molecular Biology, John Wiley and Sons (1994).
Oligonucleotide primers disclosed herein are capable of allowing accurate
assessment of ERCCl gene expression in a fixed or fixed and paraffin embedded
tissue, as well as frozen or fresh tissue. This is despite the fact that RNA derived from FPE samples is more fragmented relative to that of fresh or frozen tissue.
Thus, the methods of the invention are suitable for use in assaying ERCCl
expression levels in FPE tissue where previously there existed no way to assay
ERCCl gene expression using fixed tissues.
From the measurement of the amount of ERCCl mRNA that is expressed in
the tumor, the skilled practitioner can make a prognosis concerning clinical
resistance of a tumor to a particular genotoxin or the survivability of a patient
receiving a particular genotoxin. A platinum-based chemotherapy or a chemotherapy
inducing a similar type of DNA damage, is the preferable genotoxin.
Platinum-based chemotherapies cause a "bulky adduct" of the DNA, wherein
the primary effect is to distort the three-dimensional conformation of the double
helix. Such compounds are meant to be administered alone, or together with other
chemotherapies such as gemcitabine (Gem) or 5-Fluorouracil (5-FU).
Platinum-based genotoxic chemotherapies comprises heavy metal
coordination compounds which form covalent DNA adducts. Generally, these heavy
metal compounds bind covalently to DNA to form, in pertinent part,
cis-l,2-intrastrand dinucleotide adducts. Generally, this class is represented by
cis-diamminedichloroplatinum (II) (cisplatin), and includes cis-diammine-
(l,l-cyclobutanedicarboxylato) platinum(II) (carboplatin), cis-diammino -
(1,2-cyclohexyl) dichloroplatinum(II), and cis-(l,2-ethylenediammine)
dichloroplatinum(II). Platinum first agents include analogs or derivatives of any of
the foregoing representative compounds.
Tumors currently manageable by platinum coordination compounds include
testicular, endometrial, cervical, gastric, squamous cell, adrenocortical and small cell lung carcinomas along with medulloblastomas and neuroblastomas."
Trans-Diamminedichloroplatinum (II) (trans-DDP) is clinically useless owing, it is
thought, to the rapid repair of its DNA adducts. The use of trans-DDP as a
chemotherapeutic agent herein likely would provide a compound with low toxicity
in nonselected cells, and high relative toxicity in selected cells. In a preferred
embodiment, the platinum compound is cisplatin.
Many compounds are commonly given with platinum-based chemotherapy
agents. For example, BEP (bleomycin, etoposide, cisplatin) is used for testicular
cancer, MNAC (methotrexate, vinblastine, doxorubicin, cisplatin) is used for bladder
cancer, MNP (mitomycin C, vinblastine, cisplatin) is used for non-small cell lung
cancer treatment. Many studies have documented interactions between platinum-
containing agents. Therapeutic drug synergism, for example, has been reported for many drugs potentially included in a platinum based chemotherapy. A very short list
of recent references for this include the following: Okamoto et al., Urology 2001;
57:188-192.; Tanaka et al., Anticancer Research 2001; 21 :313-315; Slamon et al.,
Seminars in Oncology 2001; 28:13-19; Lidor et al., Journal of Clinical Investigation
1993; 92:2440-2447; Leopold et al., ΝCI Monographs 1987;99-104; Ohta et al.,
Cancer Letters 2001; 162:39-48; van Moorsel et al, British Journal of Cancer 1999;
80:981-990.
Other genotoxic agents are those that form persistent genomic lesions and are
preferred for use as chemotherapeutic agents in the clinical management of cancer.
The rate of cellular repair of genotoxin-induced DΝA damage, as well as the rate of
cell growth via the cell division cycle, affects the outcome of genotoxin therapy.
Unrepaired lesions in a cell's genome can impede DΝA replication, impair the replication fidelity of newly synthesized DNA or hinder the expression of genes
needed for cell survival. Thus, one determinant of a genotoxic agent's cytotoxicity
(propensity for contributing to cell death) is the resistance of genomic lesions
formed therefrom to cellular repair. Genotoxic agents that form persistent genomic
lesions, e.g., lesions that remain in the genome at least until the cell commits to the
cell cycle, generally are more effective cytotoxins than agents that form transient,
easily repaired genomic lesions.
A general class of genotoxic compounds that are used for treating many
cancers and that are affected by levels of ERCCl expression are DNA alkylating
agents and DNA intercalating agents. Psoralens are genotoxic compounds known to be useful in the photochemotherapeutic treatment of cutaneous diseases such as
psoriasis, vitiligo, fungal infections and cutaneous T cell lymphoma. Harrison's Principles of Internal Medicine, Part 2 Cardinal Manifestations of Disease, Ch. 60
(12th ed. 1991). Another general class of genotoxic compounds, members of which
can alkylate or intercalate into DNA, includes synthetically and naturally sourced
antibiotics. Of particular interest herein are antineoplastic antibiotics, which include
but are not limited to the following classes of compounds represented by: amsacrine;
actinomycin A, C, D (alternatively known as dactinomycin) or F (alternatively KS4);
azaserine; bleomycin; carminomycin (carubicin), daunomycin (daunorubicin), or
14-hydroxydaunomycin (adriamycin or doxorubicin); mitomycin A, B or C;
mitoxantrone; plicamycin (mithramycin); and the like.
Still another general class of genotoxic agents that are commonly used and
that alkylate DNA, are those that include the haloethylnitrosoureas, especially the
chloroethylnitrosoureas. Representative members of this broad class include carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine and
streptozotocin. Haloethylnitrosourea first agents can be analogs or derivatives of any
of the foregoing representative compounds.
Yet another general class of genotoxic agents, members of which alkylate
DNA, includes the sulfur and nitrogen mustards. These compounds damage DNA
primarily by forming covalent adducts at the N7 atom of guanine. Representative
members of this broad class include chlorambucil, cyclophosphamide, ifosfamide,
melphalan, mechloroethamine, novembicin, trofosfamide and the like.
Oligonucleotides or analogs thereof that interact covalently or noncovalently with
specific sequences in the genome of selected cells can also be used as genotoxic
agents, if it is desired to select one or more predefined genomic targets as the locus
of a genomic lesion.
Another class of agents, members of which alkylate DNA, include the
ethylenimines and methylmelamines. These classes include altretamine
(hexamethylmelamine), triethylenephosphoramide (TEPA),
triethylenethiophosphoramide (ThioTEPA) and triethylenemelamine, for example.
Additional classes of DNA alkylating agents include the alkyl sulfonates,
represented by busulfan; the azinidines, represented by benzodepa; and others,
represented by, e.g., mitoguazone, mitoxantrone and procarbazine. Each of these
classes includes analogs and derivatives of the respective representative compounds.
The invention being thus described, practice of the invention is illustrated by
the experimental examples provided below. The skilled practitioner will realize that
the materials and methods used in the illustrative examples can be modified in various ways. Such modifications are considered to fall within the scope of the
present invention.
EXAMPLES
EXAMPLE 1
RNA Isolation from FPE Tissue
RNA is extracted from paraffin-embedded tissue by the following general
procedure.
A. Deparaffinization and hydration of sections:
(1) A portion of an approximately 10 μM section is placed in a 1.5 mL
plastic centrifuge tube.
(2) 600 μL, of xylene are added and the mixture is shaken vigorously for
about 10 minutes at room temperature (roughly 20 to 25 °C).
(3) The sample is centrifuged for about 7 minutes at room temperature at the maximum speed of the bench top centrifuge (about 10-20,000 x g).
(4) Steps 2 and 3 are repeated until the majority of paraffin has been
dissolved. Two or more times are normally required depending on the amount of
paraffin included in the original sample portion.
(5) The xylene solution is removed by vigorously shaking with a lower
alcohol, preferably with 100% ethanol (about 600 μL) for about 3 minutes.
(6) The tube is centrifuged for about 7 minutes as in step (3). The
supernatant is decanted and discarded. The pellet becomes white.
(7) Steps 5 and 6 are repeated with successively more dilute ethanol
solutions: first with about 95% ethanol, then with about 80% and finally with
about 70% ethanol.
(8) The sample is centrifuged for 7 minutes at room temperature as in step
(3). The supernatant is discarded and the pellet is allowed to dry at room temperature for about 5 minutes.
B. RNA Isolation with Phenol-Chloroform
(1) 400 μL guanidine isothiocyanate solution including 0.5% sarcosine and 8
μL dithiothreitol is added.
(2) The sample is then homogenized with a tissue homogenizer (Ultra-
Turrax, IKA- Works, Inc., Wilmington, NC) for about 2 to 3 minutes while gradually
increasing the speed from low speed (speed 1) to high speed (speed 5).
(3) The sample is then heated at about 95 °C for about 5-20 minutes. It is
preferable to pierce the cap of the tube containing the sample with a fine gauge
needle before heating to 95 °C Alternatively, the cap may be affixed with a plastic
clamp or with laboratory film.
(4) The sample is then extracted with 50 μL 2M sodium acetate at pH 4.0
and 600 μL of phenol/chloroform/isoamyl alcohol (10:1.93:0.036), prepared fresh by
mixing 18 mL phenol with 3.6 mL of a 1:49 isoamyl alcoholxhloroform solution.
The solution is shaken vigorously for about 10 seconds then cooled on ice for about
15 minutes.
(5) The solution is centrifuged for about 7 minutes at maximum speed. The
upper (aqueous) phase is transferred to a new tube.
(6) The RNA is precipitated with about 10 μL glycogen and with 400
μL isopropanol for 30 minutes at -20 °C
(7) The RNA is pelleted by centrifugation for about 7 minutes in a benchtop
centrifuge at maximum speed; the supernatant is decanted and discarded; and the pellet washed with approximately 500 μL of about 70 to 75% ethanol.
(8) The sample is centrifuged again for 7 minutes at maximum speed. The supernatant is decanted and the pellet air dried. The pellet is then dissolved in an
appropriate buffer for further experiments (e.g., 50 pi. 5mM Tris chloride, pH 8.0).
EXAMPLE 2
mRNA Reverse Transcription and PCR
Reverse Transcription: RNA was isolated from microdissected or non-
microdissected formalin fixed paraffin embedded (FPE) tissue as illustrated in
Example 1 and as previously described in U.S. Application No. 09/469,338 filed
December 20, 1999, which is hereby incorporated by reference in its entirety. After
precipitation with ethanol and centrifugation, the RNA pellet was dissolved in 50 ul
of 5 mM Tris/Cl at pH 8.0. M-MLV Reverse Transcriptase will extend an
oligonucleotide primer hybridized to a single-stranded RNA or DNA template in the
presence of deoxynucleotides, producing a complementary strand. The resulting RNA was reverse transcribed with random hexamers and M-MLN Reverse
Transcriptase from Life Technologies. The reverse transcription was accomplished
by mixing 25 μl of the RΝA solution with 25.5 μl of "reverse transcription mix"
(see below). The reaction was placed in a thermocycler for 8 min at 26° C (for
binding the random hexamers to RΝA), 45 min at 42° C (for the M-MLN reverse
transcription enzymatic reaction) and 5 min at 95° C (for heat inactivation of
DΝAse).
"Reverse transcription mix" consists of 10 ul 5X buffer (250 mM Tris-HCl,
pH 8.3, 375 mM KC1, 15 mM MgC12), 0.5 ul random hexamers (50 O.D. dissolved
in 550 ul of 10 mM Tris-HCl pH 7.5) 5 ul 10 mM dΝTPs (dATP, dGTP, dCTP and dTTP), 5 ul 0.1 M DTT, 1.25 ul BSA (3mg/ml in 10 mM Tris-HCL, pH 7.5), 1.25 ul RNA Guard 24,800U/ml (RNAse inhibitor) (Porcine #27-0816, Amersham
Pharmacia) and 2.5 ul MMLV 200U/ul (Life Tech Cat #28025-02).
Final concentrations of reaction components are: 50 mM Tris-HCl, pH 8.3,
75 mM KC1, 3 mM MgC12, 1.0 mM dNTP, 1.0 mM DTT, 0*00375. mg/ml BSA,
0.62 U/ul RNA Guard and 10 U/ ul MMLV.
PCR Quantification of mRNA expression. Quantification of ERCCl
cDNA and an internal control or house keeping gene (e.g., β-actin) cDNA was done
using a fluorescence based real-time detection method (ABI PRISM 7700 or 7900
Sequence Detection System [TaqMan®], Applied Biosystems, Foster City, CA.) as
described by Heid et al, (Genome Res 1996;6:986-994); Gibson et al., (Genome Res
1996;6:995-1001). In brief, this method uses a dual labelled fluorogenic TaqMan® oligonucleotide probe, (ERCCl -53 OTc (SEQ ID NO: 3), Tm = 70° C), that anneals
specifically within the forward and reverse primers. Laser stimulation within the
capped wells containing the reaction mixture causes emission of a 3 'quencher dye
(TAMRA) until the probe is cleaved by the 5' to 3 'nuclease activity of the DNA
polymerase during PCR extension, causing release of a 5' reporter dye (6FAM).
Production of an amplicon thus causes emission of a fluorescent signal that is
detected by the TaqMan®' s CCD (charge-coupled device) detection camera, and the
amount of signal produced at a threshold cycle within the purely exponential phase
of the PCR reaction reflects the starting copy number of the sequence of interest.
Comparison of the starting copy number of the sequence of interest with the starting
copy number of theinternal control gene provides a relative gene expression level.
TaqMan® analyses yield values that are expressed as ratios between two absolute
measurements (gene of interest/internal control gene). The PCR reaction mixture consisted 0.5μl of the reverse transcription
reaction containing the cDNA prepared as described above 600 nM of each
oligonucleoride primer (ERCC1-504F (SEQ ID NO. l), Tm = 59° C and ERCC1-
574R (SEQ ID NO: 2), Tm = 58° C ), 200 nM TaqMan® probe (SEQ ID NO:3), 5 U
AmpliTaq Gold Polymerase, 200 μM each dATP, dCTP, dGTP, 400 μM dTTP, 5.5
mM MgCl2, and 1 x TaqMan® Buffer A containing a reference dye, to a final
volume of less than or equal to 25 μl (all reagents Applied Biosystems, Foster City,
CA). Cycling conditions were, 95 °C for 10 min, followed by 45 cycles at 95 °C for
15s and 60 °C for 1 min. Oligonucleotides used to quantify internal control gene β-
Actm were β-Actin TaqMan® probe (SEQ ID NO: 4), β-Actin-592F (SEQ ID NO:
5) and β-Actin-651R (SEQ ID NO: 6).
The oligonucleotide primers ERCC1-504F (SEQ ID NO:l) and ERCC1-
574R (SEQ ID NO: 2), used in the above described reaction will amplify a 71bp
product.
EXAMPLE 3
Determining the Uncorrected Gene Expression (UGE) for ERCCl
Two pairs of parallel reactions are carried out, i.e., "test" reactions and the
"calibration" reactions. The ERCCl amplification reaction and the β-actin internal
control amplification reaction are the test reactions. Separate ERCCl and β-actin
amplification reactions are performed on the calibrator RNA template and are
referred to as the calibration reactions. The TaqMan® instrument will yield four
different cycle threshold (Ct) values: Ct^^ and Ctp.act;n from the test reactions and
CtERCC1 and Ctp.oct&lfrom the calibration reactions. The differences in Ct values for the two reactions are determined according to the following equation:
ΔCttest= t^ccj - Ctp.oc(in (From the "test" reaction)
ΔCtcaIibrator= Ct^Rcc - Ctp.actin (From the "calibration" reaction)
Next the step involves raising the number 2 to the negative ΔCt, according to the following equations.
2"ΔCt tcst (From the "test" reaction)
2*ΔC'ca_ibra.or (From the "calibration" reaction)
In order to then obtain an uncorrected gene expression for ERCCl from the
TaqMan® instrument the following calculation is carried out:
Uncorrected gene expression (UGE) for ERCCl = 2 -ΔCt test / / 2 <->-ΔCt calibrator
Normalizing UGE with known relative ERCCl expression levels
The normalization calculation entails a multiplication of the UGE with a correction factor (K^cc;) specific to ERCCl and a particular calibrator RNA. A
correction factor K^^ can also be determined for any internal control gene and any
accurately pre-quantified calibrator RNA. Preferably, the internal control gene β-
actin and the accurately pre-quantified calibrator RNA Human Liver Total RNA
(Stratagene, Cat. #735017), are used. Given these reagents correction factor K^c,
equals 1.54 x 10"3.
Normalization is accomplished using a modification of the ΔCt method
described by Applied Biosystems, the TaqMan® manufacturer, in User Bulletin #2
and described above. To carry out this procedure, the UGE of 6 different test
tissues was analyzed for ERCCl expression using the TaqMan® methodology
described above. The internal control gene β-actin and the calibrator RNA,Human Liver Total RNA (Stratagene, Cat. #735017) was used.
The known relative ERCCl expression level of each sample AG221,
AG222, AG252, Adult Lung, PC3, AdCol was divided by its corresponding
TaqMan® derived UGE to yield an unaveraged correction factor K.
Kunaveraged = KllOWIl ValuβS / UGE
Next, all of the K values are averaged to determine a single K^^ correction
factor specific for ERCCl, Human Liver Total RNA (Stratagene, Cat. #735017)
from calibrator RNA and β-actin.
Therefore, to determine the Corrected Relative ERCCl Expression in an
unknown tissue sample on a scale that is consistent with pre-TaqMan® ERCCl
expression studies, one merely multiplies the uncorrected gene expression data (UGE) derived from the TaqMan® apparatus with the Kmcci specific correction
factor, given the use of the same internal control gene and calibrator RNA.
Corrected Relative ERCCl Expression = UGE x K^,-^
A K^JCC; may be determined using any accurately pre-quantified calibrator
RNA or internal control gene. Future sources of accurately pre-quantified RNA can
be calibrated to samples with known relative ERCCl expression levels as described
in the method above or may now be calibrated against a previously calibrated
calibrator RNA such as Human Liver Total RNA (Stratagene, Cat. #735017)
described above.
For example, if a subsequent K^c, is determined for a different internal
control gene and/or a different calibrator RNA, one must calibrate both the internal control gene and the calibrator RNA to tissue samples for which ERCCl expression
levels relative to that particular internal control gene have already been determined.
Such a determination can be made using standard pre-TaqMan®, quantitative RT-
PCR techniques well known in the art. The known expression levels for these
samples will be divided by their corresponding UGE levels to determine a K for that
sample. K values are then averaged depending on the number of known samples to
determine a new specific to the different internal control gene and/or
calibrator RNA.
EXAMPLE 4
All patients were enrolled in the Cisplatin/Gemcitabine arm of a prospective
multicenter three arm randomized trial (GEPC/98-02, Spanish Lung Cancer Group Phase III trial of Cisplatin/Gemcitabine (CG) versus Cisplatin/ Gemcitabine /
Ninorelbine (CGN) versus sequential doublets of Gemcitabine / Ninorelbine
followed by Ifosfamide / Ninorelbine (GNAN) in advanced ΝSCLC). All patients
received Gem 1250 mg/m2 days 1,8 plus CDDP 100mg/m2 day 1 every 3 weeks.
Eligibility criteria for GEPC/98-02 were measurable stage IV (with brain metastases
eligible if asymptomatic) or stage IIIB (malignant pleura! and/or pericardial effusion
and/or supraclavicular adenopathy) ΝSCLC and Eastern Cooperative Group (ECOG)
performance score 0-2. All patients had chest x-ray and a computed tomography
(CT) scan of the chest and upper abdomen before entry into the study and underwent
repeat evaluations at least every 6 weeks. Tumor response was assessed according to
WHO criteria as complete response, partial response, stable disease, and progressive disease. Tumors were reassessed during treatment with the same imaging methods
used to establish the baseline tumor measurement.
Total mRNA was isolated from microdissected FPE pretreatment tumor
samples, and Corrected Relative ERCCl Expression was measured using
quantitative RT-PCR as described in Examples 2 and 3. A method for mRNA
isolation from such samples is described in Example 1 and in US Patent Application
No. 09/469,338, filed December 20, 1999, and is hereby incorporated by reference in
its entirety.
Statistical Analysis
The Mann- Whitney U test was used to test for significant associations
between the continuous test variable Corrected Relative ERCCl Expression and
dichotomous variables (patient sex, age above and below the median age, presence of weight loss, presence of pleural effusion, tumor stage). The Kruskal-Wallis test
was used to test for significant differences in Corrected Relative ERCCl Expression
within multiple groups (ECOG performance status, histopathology). Fisher's exact
test was used for the analysis of categorical clinicopatho logical values including
response and dichotomized Corrected Relative ERCCl Expression values.
All patients were followed from first study treatment until death or until the
data were censored. Kaplan-Meier survival curves and the log rank test were used to
analyze univariate distributions for survival and disease-free survival. The maximal
chi-square method of Miller and Siegmund (Biometrics 1982; 38:1011-1016 and
Halpera (Biometrics 1982; 38:1017-1023) was adapted to determine which
expression value best segregated patients into poor- and good prognosis subgroups (in terms of likelihood of surviving), with the log-rank test as the statistic used to
measure the strength of the grouping. To determine a P value that would be
interpreted as a measure of the strength of the association based on the maximal chi-
square analysis, 1000 boot-strap-like simulations were used to estimate the
distribution of the maximal chi-square statistics under the hypothesis of no
association.(Biometrics 1982; 38: 1017-1023) Cox's proportional hazards modeling
of factors that were significant in univariate analysis was performed to identify
which factors might have a significant influence on survival. SPSS version 10.0.5
software (SPSS Inc., Chicago 111.) was used for all statistical analyses. All P values were
two-sided.
Corrected Relative ERCCl Expression Levels.
ERCCl mRNA expression was detectable in all 56 samples analyzed. The
median Corrected Relative ERCCl Expression, relative to the expression of the
internal control housekeeping gene β-actin, was 6.7 x 10"3 (range 0.8 x 10"3 - 24.6 x
10"3). There were no significant associations between Corrected Relative ERCCl
Expression levels and any of the factors age (P= 0.66), sex (P=0.18) presence of
weight loss in the six months prior to randomization (P=0.74), tumor stage (IIIB
versus IN, P=0.39), or presence of pleural effusion (P=0.25, all Mann- Whitney U
test). There were also no significant differences between the Corrected Relative
ERCCl Expression levels among patients with different performance status grades
(P=0.48, Kruskal-Wallis test) or different tumor cell types (all four tumor types,
P=0.10, Kruskal-Wallis test), but Corrected Relative ERCCl Expression levels were significantly higher in SCC tumors (median 8.6 x 10"3) compared to
adenocarcinomas (median 5.2 x 10"3, P=0.015, Mann- Whitney test).
Response to chemotherapy
The tumor response frequencies for the 47 patients who were evaluable for
response are shown in Figure 3. The overall response rate was 44.7%. The Corrected
Relative ERCCl Expression levels in the complete response and partial response i.e. "responding" tumors (median 4.3 x 10"3, range 1.2 x 10"3-24.6 x 10"3) were not
significantly different from the levels in the stable disease and progressive disease
i.e. "non-responding" tumors (median 7.85 x 10"3, range 0.8 x 10"3-24.3 x 10"3,
P=0.31 Mann- Whitney test). There were also no significant differences between the
proportion of responding and non-responding tumors with Corrected Relative
ERCCl Expression values greater and less than any ERCCl level (all Fisher's exact
test). The response rate in tumors with Corrected Relative ERCCl Expression below
the threshold value ("low" expression, 52% responders) was higher than for tumors
with Corrected Relative ERCCl Expression above the threshold value ("high"
expression, 36.4% responders, Fisher's exact test, P = 0.38).
Association between patient overall survival and Corrected Relative ERCCl
Expression levels
The median overall survival time was 36.6 weeks (range 0-113.4 weeks) and
the median time to progression was 24.4 weeks (range 0-102.9 weeks). Use of the
log rank test and the maximal chi-square statistic to identify threshold Corrected
Relative ERCCl Expression levels that segregated patients into poor- and good- prognosis subgroups showed that the range of discriminatory values included the
median value, which was therefore used as the threshold value for the survival
analysis. Therefore, the threshold Corrected Relative ERCCl Expression value was
determined to be 6.7 x 10"3 for NSCLC Figure 1 shows the Kaplan-Meier survival
curve for patients with intratumoral Corrected Relative ERCCl Expression levels
above and below the threshold Corrected Relative ERCCl Expression level. As
shown in Figure 4, patients with Corrected Relative ERCCl Expression levels below
the threshold value had a significantly longer median survival of 61.6 weeks (95%
CI. 42.4, 80.7 weeks) compared to 20.4 weeks (95% CI. 6.9, 33.9 weeks) for
patients with Corrected Relative ERCCl Expression levels above the threshold
value. Adjusted for tumor stage, the log rank statistic for the association between
low or high Corrected Relative ERCCl Expression and overall survival was 3.97 and the P value was 0.046. The unadjusted log rank results are shown in Figure 4.
A separate Corrected Relative ERCCl Expression threshold value of 5.8 x
10"3 was tested because this value was shown in a previous study to be associated
with overall survival for patients with gastric cancer. (Metzger et al., J Clin Oncol
1998; 16:309-316). Overall survival was significantly better for the group of NSCLC
patients in this study with Corrected Relative ERCCl Expression levels less than 5.8
x 10"3 compared to those with ERCCl levels less than 5.8 x 10"3 (log rank statistic
6.37, P=0.011), although a higher 6.7 x 10"3 Corrected Relative ERCCl Expression
threshold level is a more powerful discriminator.
Other factors that were significantly associated with overall survival on
univariable analysis using Kaplan Meier survival curves and the log rank test were the presence of pretreatment weight loss and the ECOG performance status (Table 2). Patient age (P=0.18), sex (P=0.87), tumor stage (P=0.99), tumor cell type
(P=0.63), and presence of pleural effusion (P=0.71) were not significant prognostic
factors for overall survival. Corrected Relative ERCCl Expression level, ECOG
performance status, and weight loss remained significant prognostic factors for
survival in the Cox proportional hazards regression model multivariable analysis
(Figure 4). P values for a Cox regression model stratified on tumor stage were 0.038
for ERCCl, 0.017 for weight loss, and 0.02 for ECOG performance status (PS 0
versus 1 or 2).
This study found an association between lower ERCCl mRNA expression
levels and improved survival after treatment with a platinum-based
chemotherapeutic for patients with cancer.

Claims (14)

1. A method for determining a platinum-based chemotherapeutic
regimen for treating a tumor in a patient comprising:
(a) obtaining a tissue sample of the tumor and fixing the sample, to
obtain a fixed tumor sample;
(b) isolating mRNA from the fixed tumor sample;
(c) subjecting the mRNA to amplification using a pair of oligonucleotide
primers that hybridize under stringent conditions to a region of the
ERCCl gene, to obtain an amplified sample; (d) determining the amount of ERCCl mRNA in the amplified sample;
(e) comparing the amount of ERCCl mRNA from step (d) to an amount
of mRNA of an internal control gene; and
(f) determining a platinum-based chemotherapeutic regimen based on
the amount of ERCCl mRNA in the amplified sample and a
predetermined threshold level for ERCCl gene expression.
2. The method of claim 1 wherein the oligonucleotide primers consist of the
oligonucleotide primer pair ERCCl, or pair of oligonucleotide primers
substantially identical thereto.
3. The method of claim 1 wherein, the tumor is a non-small-cell lung cancer
(NSCLC) tumor.
4. The method of claim 1 wherein, the threshold level of ERCCl gene
expression is about 6.7 x 10"3 times internal control gene expression level.
5. A method of treating a tumor with a platinum-based chemotherapeutic
regimen comprising:
(a) obtaining a tissue sample of the tumor and fixing the sample,
to obtain a fixed tumor sample;
(b) isolating mRNA from the fixed tumor sample;
(c) subjecting the mRNA to amplification using a pair of
oligonucleotide primers that hybridize under stringent
conditions to a region of the ERCCl gene, to obtain an amplified sample;
(d) determining the amount of ERCCl mRNA in the amplified sample;
(e) comparing the amount of ERCCl mRNA from step (d) to an
amount of mRNA of an internal control gene;
(f) providing a platinum-based chemotherapeutic regimen
comprising a genotoxic agent when the determined gene
expression level for ERCCl gene is below a predetermined threshold value.
6. The method of claim 5 wherein, the tumor is a non-small-cell lung cancer (NSCLC) tumor.
7. The method of claim 5 wherein, the genotoxic agent is gemcitabine, cisplatin
or a combination thereof.
8. A method for determining the level of ERCCl expression in a fixed paraffin
emedded tissue sample comprising;
(a) deparaffinizing the tissue sample; to obtain a deparaffinized sample;
(b) isolating mRNA from the deparaffinized sample in the presence of an
effective amount of a chaotropic agent;
(c) subjecting the mRNA to amplification using a pair of oligonucleotide
primers that hybridize under stringent conditions to a region of the ERCCl gene, to obtain an amplified sample;
(d) determining the quantity of ERCCl mRNA relative to the quantity of an internal control gene's mRNA.
9. The method of claim 8 wherein, the pair of oligonucleotide primers consists
of the oligonucleotide primer pair ERCCl or a pair of oligonucleotide
primers substantially similar thereto.
10. The method of claim 8 wherein, the internal control gene is β-actin.
11. The method of claim 8 wherein, mRNA isolation is carried out by
(a) heating the tissue sample in a solution comprising an effective
concentration of a chaotropic compound to a temperature in the range of about 75 to about 100 °C for a time period of about 5 to about 120
minutes;
and
(b) recovering said mRNA from said chaotropic solution.
12. An oligonucleotide primer having the sequence of SEQ ID NO: 1 or and an
oligonucleotide substantially identical thereto.
13. An oligonucleotide primer having the sequence of SEQ ID NO: 2 or and an
oligonucleotide substantially identical thereto.
14. A kit for detecting expression of an ERCCl gene comprising,
oligionucleotide pair ERCCl or an oligonucleotide pair substantially identical thereto.
AU2002249768A 2000-12-01 2001-11-29 Method of determining a chemotherapeutic regimen based on ERCC1 expression Ceased AU2002249768B2 (en)

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
US25012100P 2000-12-01 2000-12-01
US60/250,121 2000-12-01
US25047000P 2000-12-04 2000-12-04
US60/250,470 2000-12-04
US09/796,491 US6518416B1 (en) 2000-12-01 2001-03-02 Method of determining a chemotherapeutic regimen based on ERCC1 expression
US09/796,491 2001-03-02
US09/877,095 2001-06-11
US09/877,095 US6573052B2 (en) 2000-12-01 2001-06-11 Method of determining a chemotherapeutic regimen based on ERCCI expression
US09/988,784 US6602670B2 (en) 2000-12-01 2001-11-20 Method of determining a chemotherapeutic regimen based on ERCC1 expression
US09/988,784 2001-11-20
PCT/US2001/044519 WO2002061128A2 (en) 2000-12-01 2001-11-29 Method of determining a chemotherapeutic regimen based on ercc1 expression

Publications (2)

Publication Number Publication Date
AU2002249768A1 true AU2002249768A1 (en) 2003-02-20
AU2002249768B2 AU2002249768B2 (en) 2007-01-04

Family

ID=45773848

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2002249768A Ceased AU2002249768B2 (en) 2000-12-01 2001-11-29 Method of determining a chemotherapeutic regimen based on ERCC1 expression

Country Status (14)

Country Link
US (2) US6602670B2 (en)
EP (1) EP1364054B1 (en)
JP (1) JP4503922B2 (en)
KR (1) KR100966271B1 (en)
CN (1) CN1636069B (en)
AR (1) AR031455A1 (en)
AU (1) AU2002249768B2 (en)
CA (1) CA2437044C (en)
ES (1) ES2439873T3 (en)
HK (1) HK1078328A1 (en)
IL (2) IL156272A (en)
MX (1) MXPA03004930A (en)
NZ (1) NZ526710A (en)
WO (1) WO2002061128A2 (en)

Families Citing this family (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003534292A (en) * 2000-05-19 2003-11-18 ジェネンテック・インコーポレーテッド ERBB antagonist gene detection assays to increase the likelihood of effective response to cancer treatment
US6602670B2 (en) * 2000-12-01 2003-08-05 Response Genetics, Inc. Method of determining a chemotherapeutic regimen based on ERCC1 expression
US7049059B2 (en) * 2000-12-01 2006-05-23 Response Genetics, Inc. Method of determining a chemotherapeutic regimen based on ERCC1 and TS expression
EP3115470B1 (en) 2002-03-13 2018-07-18 Genomic Health, Inc. Gene expression profiling in biopsied tumor tissues
EP2385139A1 (en) * 2002-07-31 2011-11-09 University of Southern California Polymorphisms for predicting disease and treatment outcome
AU2003290925A1 (en) 2002-11-14 2004-06-15 John Wayne Cancer Institute Detection of micro metastasis of melanoma and breast cancer in paraffin-embedded tumor draining lymph nodes by multimaker quantitative rt-pcr
US8039218B2 (en) * 2002-11-14 2011-10-18 John Wayne Cancer Institute Detection of cancer cells in body fluids
JP4606879B2 (en) * 2002-11-15 2011-01-05 ジェノミック ヘルス, インコーポレイテッド Gene expression profiling of EGFR positive cancer
US8580498B2 (en) * 2002-11-15 2013-11-12 University Of South Florida Predictive value of nuclear excision repair genes for cancer survival
US20040231909A1 (en) 2003-01-15 2004-11-25 Tai-Yang Luh Motorized vehicle having forward and backward differential structure
EP1590487A2 (en) * 2003-02-06 2005-11-02 Genomic Health, Inc. Gene expression markers for response to egfr inhibitor drugs
JP4568716B2 (en) * 2003-02-20 2010-10-27 ジェノミック ヘルス, インコーポレイテッド Use of intron RNA to measure gene expression
CA2527285A1 (en) * 2003-05-28 2004-12-23 Genomic Health, Inc. Gene expression markers for predicting response to chemotherapy
WO2004111273A2 (en) * 2003-05-30 2004-12-23 Genomic Health, Inc. Gene expression markers for response to egfr inhibitor drugs
EP3470535B1 (en) 2003-06-24 2020-04-01 Genomic Health, Inc. Prediction of likelihood of cancer recurrence
WO2005008213A2 (en) * 2003-07-10 2005-01-27 Genomic Health, Inc. Expression profile algorithm and test for cancer prognosis
WO2005064019A2 (en) 2003-12-23 2005-07-14 Genomic Health, Inc. Universal amplification of fragmented rna
ES2550614T3 (en) * 2004-04-09 2015-11-11 Genomic Health, Inc. Gene expression markers to predict the response to chemotherapy
AU2005267148A1 (en) * 2004-07-01 2006-02-02 University Of Southern California Genetic markers for predicting disease and treatment outcome
CA3061785A1 (en) * 2004-11-05 2006-05-18 Genomic Health, Inc. Predicting response to chemotherapy using gene expression markers
WO2006052731A2 (en) 2004-11-05 2006-05-18 Genomic Health, Inc. Molecular indicators of breast cancer prognosis and prediction of treatment response
US7449184B2 (en) 2005-01-21 2008-11-11 Genentech, Inc. Fixed dosing of HER antibodies
AU2006213659A1 (en) * 2005-02-09 2006-08-17 Genentech, Inc. Inhibiting HER2 shedding with matrix metalloprotease antagonists
PT1850874E (en) 2005-02-23 2013-12-02 Genentech Inc Extending time to disease progression or survival in ovarian cancer patients using pertuzumab
WO2006096861A2 (en) * 2005-03-08 2006-09-14 Genentech, Inc. METHODS FOR IDENTIFYING TUMORS RESPONSIVE TO TREATMENT WITH HER DIMERIZATION INHIBITORS (HDIs)
EP2460886B1 (en) * 2005-05-04 2014-01-01 University of South Florida Predicting treatment response in cancer subjects
JP2006316040A (en) 2005-05-13 2006-11-24 Genentech Inc Herceptin(r) adjuvant treatment
US9702875B2 (en) 2006-03-14 2017-07-11 Institute Gustave-Roussy Expression of isoform 202 of ERCC1 for predicting response to cancer chemotherapy
EP1835285A1 (en) * 2006-03-14 2007-09-19 Institut Gustave Roussy ERCC1 expression in predicting response for cancer chemotherapy
PE20090681A1 (en) 2007-03-02 2009-06-10 Genentech Inc PREDICTION OF RESPONSE TO A HER INHIBITOR
US9551033B2 (en) * 2007-06-08 2017-01-24 Genentech, Inc. Gene expression markers of tumor resistance to HER2 inhibitor treatment
CA2690334C (en) 2007-06-08 2017-02-14 Genentech, Inc. Gene expression markers of tumor resistance to her2 inhibitor treatment
EP2171086B1 (en) * 2007-06-15 2012-12-19 University of South Florida Methods of diagnosing and treating cancer
CA2700978A1 (en) * 2007-09-28 2009-04-02 Duke University Individualized cancer treatments
CN101140241B (en) * 2007-09-30 2010-11-03 浙江省肿瘤医院 Platinum kind anti-cancer drugs susceptibility related gene inspect reagent kit and applications thereof
BRPI0812682A2 (en) 2008-06-16 2010-06-22 Genentech Inc metastatic breast cancer treatment
WO2010091763A1 (en) * 2009-02-16 2010-08-19 Atlas Antibodies Ab Rbm3 as a marker for malignant melanoma prognosis
ES2572728T3 (en) 2009-03-20 2016-06-02 F. Hoffmann-La Roche Ag Bispecific anti-HER antibodies
JP5705836B2 (en) 2009-05-29 2015-04-22 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト Modulators for HER2 signaling in gastric cancer patients expressing HER2
US9556249B2 (en) 2010-02-18 2017-01-31 Genentech, Inc. Neuregulin antagonists and use thereof in treating cancer
WO2011146568A1 (en) 2010-05-19 2011-11-24 Genentech, Inc. Predicting response to a her inhibitor
JP5769952B2 (en) * 2010-11-12 2015-08-26 株式会社Lsiメディエンス Highly sensitive detection method for EML4-ALK fusion gene
US20130245233A1 (en) 2010-11-24 2013-09-19 Ming Lei Multispecific Molecules
CA2819080A1 (en) * 2010-12-09 2012-06-14 Juan De La Haba-Rodriguez Agtr1 as a marker for bevacizumab combination therapies
EP2655413B1 (en) 2010-12-23 2019-01-16 F.Hoffmann-La Roche Ag Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
US9626650B2 (en) 2011-04-14 2017-04-18 Elwha Llc Cost-effective resource apportionment technologies suitable for facilitating therapies
US10445846B2 (en) 2011-04-14 2019-10-15 Elwha Llc Cost-effective resource apportionment technologies suitable for facilitating therapies
US8599009B2 (en) 2011-08-16 2013-12-03 Elwha Llc Systematic distillation of status data relating to regimen compliance
KR20140057326A (en) 2011-08-17 2014-05-12 제넨테크, 인크. Neuregulin antibodies and uses thereof
WO2013063229A1 (en) 2011-10-25 2013-05-02 The Regents Of The University Of Michigan Her2 targeting agent treatment in non-her2-amplified cancers having her2 expressing cancer stem cells
RU2019103083A (en) 2011-11-30 2019-03-22 Дженентек, Инк. Mutations of ErbB3 in Cancer
US9376715B2 (en) 2011-12-09 2016-06-28 Roche Molecular Systems, Inc Methods for detecting mutations in the catalytic subunit of the phosphoinositol-3 kinase (PIK3CA) gene
US10679309B2 (en) 2011-12-30 2020-06-09 Elwha Llc Evidence-based healthcare information management protocols
US10552581B2 (en) 2011-12-30 2020-02-04 Elwha Llc Evidence-based healthcare information management protocols
US10528913B2 (en) 2011-12-30 2020-01-07 Elwha Llc Evidence-based healthcare information management protocols
US10559380B2 (en) 2011-12-30 2020-02-11 Elwha Llc Evidence-based healthcare information management protocols
US10475142B2 (en) 2011-12-30 2019-11-12 Elwha Llc Evidence-based healthcare information management protocols
US10340034B2 (en) 2011-12-30 2019-07-02 Elwha Llc Evidence-based healthcare information management protocols
US20130173298A1 (en) 2011-12-30 2013-07-04 Elwha LLC, a limited liability company of State of Delaware Evidence-based healthcare information management protocols
EP2831115A1 (en) 2012-03-27 2015-02-04 F. Hoffmann-La Roche AG Diagnosis and treatments relating to her3 inhibitors
CN102676661B (en) * 2012-04-27 2014-12-31 中国人民解放军第四军医大学 Fluorescent polarization based homogeneous phase detection method of single nucleotide polymorphism of codon118 of ERCC1 (excision repair cross-complementing 1) gene
US10450609B2 (en) * 2012-05-22 2019-10-22 The Johns Hopkins University Quantitative multiplex methylation specific PCR method—cMethDNA, reagents, and its use
BR112015012644A2 (en) 2012-11-30 2017-12-19 Hoffmann La Roche method for determining a cancer patient's need, cancer treatment method, pharmaceutical composition, use of a nucleic acid or antibody and kit;
EP3014505A4 (en) * 2013-06-28 2017-03-08 Nantomics, LLC Pathway analysis for identification of diagnostic tests
GB201316024D0 (en) * 2013-09-09 2013-10-23 Almac Diagnostics Ltd Molecular diagnostic test for lung cancer
CN103451303B (en) * 2013-09-12 2015-02-11 江苏康克生物技术有限公司 Kit for detecting expression level of human ERCC1 (excision repair cross complementation 1) through PCR (polymerase chain reaction) method
CN105219881B (en) * 2015-11-17 2018-11-20 东南大学 The kit of quantitative detection ERCC1-202 a kind of and application
EP3454863A1 (en) 2016-05-10 2019-03-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Combinations therapies for the treatment of cancer
CN110268269A (en) * 2016-12-18 2019-09-20 赛隆特拉有限公司 Using the gene that APOE4 motif mediates to be used for diagnosing and treating Alzheimer's disease
KR20200019227A (en) * 2017-06-20 2020-02-21 난토믹스, 엘엘씨 USE OF CELL-FREE CIRCULATING RNA (cfRNA) EXPRESSION OF PD-L1 AND ERCC1 IN PLASMA TO MONITOR RESPONSE TO THERAPY IN NSCLC)

Family Cites Families (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4830969A (en) 1981-08-31 1989-05-16 The Research Foundation Of State University Of New York Process for the rapid and simple isolation of nucleic acids
US4843155A (en) 1987-11-19 1989-06-27 Piotr Chomczynski Product and process for isolating RNA
US5128247A (en) 1989-08-14 1992-07-07 Board Of Regents, The University Of Texas System Methods for isolation of nucleic acids from eukaryotic and prokaryotic sources
US5654179A (en) 1990-11-14 1997-08-05 Hri Research, Inc. Nucleic acid preparation methods
US5284940A (en) 1990-11-14 1994-02-08 Hri Research, Inc. Preparation for nucleic acid samples
US5620852A (en) 1990-11-14 1997-04-15 Hri Research, Inc. Nucleic acid preparation methods
JP3171863B2 (en) * 1991-01-16 2001-06-04 伊藤製油株式会社 Amphoteric surfactant composition
US5346994A (en) 1992-01-28 1994-09-13 Piotr Chomczynski Shelf-stable product and process for isolating RNA, DNA and proteins
US5502166A (en) 1992-02-26 1996-03-26 Mitsubishi Chemical Corporation NMDH receptor proteins and genes encoding the same
WO1994018156A1 (en) 1993-02-01 1994-08-18 University Of Iowa Research Foundation Quartenary amine surfactants and methods of using same in isolation of rna
US5637687A (en) 1993-08-31 1997-06-10 Wiggins; James C. Methods and compositions for isolating nucleic acids
AU2289495A (en) 1994-04-13 1995-11-10 Uab Research Foundation, The Dihydropyrimidine dehydrogenase compositions and methods of use
US5643767A (en) 1994-05-02 1997-07-01 The Rockefeller University Process for isolating cellular components
CA2153215A1 (en) 1994-07-06 1996-01-07 Lu Wang Treatment of paraffin embedded tissue for gene analysis
JP3125633B2 (en) * 1994-07-06 2001-01-22 和光純薬工業株式会社 Method and kit for processing paraffin-embedded tissue specimen as sample for gene analysis
US5707802A (en) 1995-01-13 1998-01-13 Ciba Corning Diagnostics Corp. Nucleic acid probes for the detection and identification of fungi
US5777099A (en) 1995-02-24 1998-07-07 Biotecx Laboratories, Inc. RNA separation
US5705336A (en) * 1995-03-07 1998-01-06 The United States Of America As Represented By The Department Of Health And Human Services Assay for sensitivity of tumors to DNA-platinating chemotherapy
US5945515A (en) 1995-07-31 1999-08-31 Chomczynski; Piotr Product and process for isolating DNA, RNA and proteins
DE29601618U1 (en) 1996-01-31 1996-07-04 Invitek Gmbh Multiple simultaneous isolation device
EP0791654A1 (en) 1996-02-21 1997-08-27 Jürgen A. Dr. Richt Polypeptides corresponding to the amino acid sequences of proteins p57 or p9.5 of Borna disease virus, nucleic acid fragments coding therefore and their use for diagnostic and immunization purposes
DK0896629T3 (en) 1996-03-20 2003-02-03 Us Gov Health & Human Serv Methods and compositions for detecting splicing defects in the dihydropyrimidine dehydrogenase gene
CA2253937A1 (en) 1996-05-10 1997-11-20 Phylomed Corporation Methods for oxidizing disulfide bonds using ozone
AU6764398A (en) 1997-03-20 1998-10-12 Variagenics, Inc. Target genes for allele-specific drugs
US5994076A (en) 1997-05-21 1999-11-30 Clontech Laboratories, Inc. Methods of assaying differential expression
DK1005633T3 (en) 1997-08-20 2008-12-15 Univ Miami Tissue fixation dehydration fat removal impregnation method with high quality and continuous flow
US5952202A (en) 1998-03-26 1999-09-14 The Perkin Elmer Corporation Methods using exogenous, internal controls and analogue blocks during nucleic acid amplification
US6204375B1 (en) 1998-07-31 2001-03-20 Ambion, Inc. Methods and reagents for preserving RNA in cell and tissue samples
US6248535B1 (en) 1999-12-20 2001-06-19 University Of Southern California Method for isolation of RNA from formalin-fixed paraffin-embedded tissue specimens
US7049059B2 (en) * 2000-12-01 2006-05-23 Response Genetics, Inc. Method of determining a chemotherapeutic regimen based on ERCC1 and TS expression
US6602670B2 (en) * 2000-12-01 2003-08-05 Response Genetics, Inc. Method of determining a chemotherapeutic regimen based on ERCC1 expression

Similar Documents

Publication Publication Date Title
CA2437044C (en) Method of determining a chemotherapeutic regimen based on ercc1 expression
AU2002249768A1 (en) Method of determining a chemotherapeutic regimen based on ERCC1 expression
US7560543B2 (en) Method of determining a chemotherapeutic regimen based on ERCC1 and TS expression
AU2002246504A1 (en) Method of determining a chemotherapeutic regimen based on ERCC1 and TS expression
JP2009000118A (en) METHOD OF DETERMINING CHEMOTHERAPY BASED ON GLUTATHIONE-S-TRANSFERASE Pi EXPRESSION
US6573052B2 (en) Method of determining a chemotherapeutic regimen based on ERCCI expression
AU2002305573A1 (en) Method of determining a chemotherapeutic regimen based on glutathione-s-transferase pi expression
AU2007203027A1 (en) Method of determining a chemotherapeutic regimen based on ERCC1 and TS expression