WO2017070497A1 - Methods and compositions for use of driver mutations in cll - Google Patents
Methods and compositions for use of driver mutations in cll Download PDFInfo
- Publication number
- WO2017070497A1 WO2017070497A1 PCT/US2016/058164 US2016058164W WO2017070497A1 WO 2017070497 A1 WO2017070497 A1 WO 2017070497A1 US 2016058164 W US2016058164 W US 2016058164W WO 2017070497 A1 WO2017070497 A1 WO 2017070497A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- del
- cll
- mutations
- subject
- cnv
- Prior art date
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/68—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
- C12Q1/6876—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
- C12Q1/6883—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
- C12Q1/6886—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
Definitions
- the present invention provides methods for treatment and prognosis of chronic lymphocytic leukemia (CLL) using novel cancer driver mutations.
- CLL chronic lymphocytic leukemia
- composition of the cohort of prior WES studies has limited the ability to accurately determine the impact of drivers and clonal heterogeneity on clinical outcome since they included samples collected at variable times from subjects exposed to a variety of therapies. Understanding the genetic alterations that drive tumorigenesis and how they evolve over the course of disease and therapy are central questions in cancer biology. Thus, identifying novel cancer drivers is necessary to improve treatment and diagnosis of CLL.
- Applicants analyzed WES data from 538 CLLs, including 278 pre- treatment samples collected from subjects enrolled on the phase III CLL8 study 13 .
- This trial established the combination of fludarabine (F), cyclophosphamide (C) and rituximab (R) as the current standard-of-care first-line treatment for patients of good physical fitness, with a median of >6 years of follow-up.
- Applicants discovered novel CLL cancer genes, performed a comprehensive genetic characterization of samples from patients prior to exposure to a uniform and contemporary treatment, and uncovered features contributing to relapse from this therapy.
- the present invention provides for a method of treating chronic lymphocytic leukemia (CLL) in a subject in need thereof comprising identifying the presence of gene mutations and somatic copy number variations (CNV) in a genomic DNA sample obtained from the subject, wherein the mutated genes and somatic copy number variations (CNV) comprise del(13q), SF3B1, A TM, del(llq), tril2, TP53, NOTCH 1, POT1, amp(2p), del(17p), CHD2, XPOl, RPS15, BIRC3, BRAF, IGLL5, d l(8p), MGA, MYD88, del(6q21), EGR2, FBXW7, ampfSq), DDX3X, KRAS, BCOR, IKZF3, MAP2K1, ZMYM3, IRF4, SAMHD1, BAZ2A, CARD!
- CLL chronic lymphocytic leukemia
- the patient may have stage 0, 1, II, III, or IV CLL.
- the method is performed before a patient has symptoms. The method may be performed before treatment, during treatment, or after treatment.
- the method may be performed when the patient is in remission.
- the presence of mutations in NRAS, KRAS, BRAF, and/or MAP2K1 may indicate treating with an inhibitor of the MAPK-ERK pathway.
- the mutation in BRjiF may be a mutation that is not the canonical V600E mutation. In this instance the treatment may include a MEK inhibitor and would not indicate the use of a BRAF inhibitor.
- the presence of mutations in RPS15 may indicate an adverse outcome and the subject is treated with an alternative treatment regimen.
- the presence of mutations in IKZF3 may indicate an adverse outcome and the subject is treated with an alternative treatment regimen.
- the mutation in IKZF3 may be an L162R substitution.
- the presence of mutations in delflSq), MYD88, and/ or ( 7/7/2 may indicate decreased clinical aggressiveness.
- the presence of mutations in TP53, SF3B1 and/or XPOl may indicate shorter progression-free survival (PFS) and overall survival (OS).
- the genomic DNA sample may be obtained prior to treatment by any methods known in the art. Not being bound by a theory, obtaining the sample prior to treatment allows the detection of the mutations or alterations in copy number of the present invention in clonal and subclonal populations of cells present in a cancer cell fraction (CCF) before treatment. Identification of mutations or alterations in copy number can be used to predict resistence to a treatment and efficacy of a treatment.
- the method may further comprising determining mutations in IGHV, wherein the presence of unmutated IGHV indicates increased clinical aggressiveness and the presence of mutated IGHV indicates decreased clinical aggressiveness.
- the present invention provides for a method of determining whether a subject having chronic lymphocytic leukemia (CLL) would derive a clinical benefit of early treatment comprising determining the presence of gene mutations and somatic copy number variations (CNV) in a genomic DNA sample obtained from the subject, wherein the mutated genes and somatic copy number variations (CNV) comprise del(13q), SF3B1, ATM, del(llq), trill, TPS 3, NOTCH 1, POT1, amp(2p), del(17p), CHD2, XPOl, RPS15, BIRC3, BRAF, IGLL5, del(8p), MGA, MYD88, del(6q21), EGR2, FBXW7, amp(8q), DDX3X, KRAS, BCOIi IKZF3, MAP2K1, 7MYM3, IRF4, SAMHD!, BAZ2A, CARDIl, FUBP1, HISTIH!E,
- BRCC3, EWSR1, FAM50A, TRAF3, ASXL1, CHEK2, GNBl, HISTIHIB, and P!Ml wherein the presence of a mutated gene and/or somatic copy number variation (CNV) indicates that the subject would derive a clinical benefit of early treatment
- the present invention provides for a method of predicting survivability of a subject having chronic lymphocytic leukemia (CLL) comprising determining the presence of gene mutations and somatic copy number variations (CN V) in a genomic DN A sample obtained from the subject, wherein the mutated genes and somatic copy number variations (CNV) co pnsQdeI(I3q), SF3B1, ATM, del(l lq), trill, TP53, NOTCH!, POT1, amp(2p), del(17p), CIID2, XPOl, RPS15, BIRC3, BRAF, IGLL5, del(8p), MGA, MYD88, del(6q21), EGR2, FBXW7, amp(8q), DDX3X, KRAS, BCOR, IKZF3, MAP2K1, 7MYM3, IRF4, SAMHD!, BAZ2A, CARDIl, FlJBPl, HI
- CLL chronic lympho
- the present invention provides for a method of identifying a candidate subject for a clinical trial for a treatment protocol for chronic lymphocytic leukemia (CLL) comprising determining the presence of gene mutations and somatic copy number variations (CNV) in a genomic DNA sample obtained from the subject, wherein the mutated genes and somatic copy number variations (CNV) comprise del(13q), SF3B1, ATM, del/1 Iq), trill, TP 53, NOTCH!, POT I, amp(lp), deI(T7p), CHD2, XPOl, RPS15, B1RC3, BRAF, IGLL5, del(8p), MGA, MYD88, deMqll), EGR2, FBXW7, mpfSq), DDX3X, KRAS, BCOR, IKZF3, MAP1K1, ZMYM3, IRF4, SAMHD1, BAZ1A, CARDll, FUBPl, H1ST1H1E,
- the present invention provides for a method of detecting chronic lymphocytic leukemia (CLL) in a subject comprising determining the presence of gene mutations and somatic copy number variations (CNV) in a genomic DNA sample obtained from the subject, wherein the mutated genes and somatic copy number variations (CNV) comprised?/ ⁇ ), SF3B1, Am, del/1 Iq), tnl2, TP 53, NOTCH 1, POT1, amp(2p), del(17p), CHD2, XPOl, RPSI5, BIRC3, BRAF, IGLL5, del(8p), MGA, MYD88, del(6q2l), EGR2, FBXW7, ⁇ , DDX3X, KRAS, BCOR, IKZF3, MAP2K1, ZMYM3, IRF4, SAMHD1, BAZ2A, CARDll, FUBPl, HISTIHIE, MED 12, NRAS, NXFI, del
- CLL chronic lympho
- the present invention provides for a method of identifying a subject at elevated risk of having CLL with rapid disease progression comprising: (a) analyzing genomic DNA in a sample obtained from a subject having or suspected of having chronic lymphocytic leukemia (CLL) for the presence of gene mutations and somatic copy number variations (CNV), wherein the mutated genes and somatic copy number variations (CNV) comprise£fe/ ⁇ 733 ⁇ 4), .SV- ' .?/>7, A TM, del(llq), tril2, TP53, NOTCH 1, POT I, amp(lp), del(l7p), CHDl, XPOl, RPS15, BIRC3, BRAF, IGLL5, del(8p), MGA, MYD88, deUfiqll), EGRl, FBXW7, amp(8q), DDX3X, KRAS, BCOR, IKZF3, MA 2 h i, ZMYM3, T
- the method may further comprise treating a subject for CLL identified as a subject at elevated risk of having CLL with rapid disease progression.
- Any of the methods may include determining mutations in more than one risk allele. Any of the methods may be repeated every month, 2 months, 3 months, 4 months, five months, 6 months 8 months, or one year or at any time where there is a change in clinical status.
- the genomic DNA sample may be obtained from peripheral blood, bone marrow, or lymph node tissue.
- the genomic DNA may be analyzed using whole genome sequencing (WGS), whole exome sequencing (WES), single nucleotide polymorphism (SNP) analysis, deep sequencing, targeted gene sequencing, hybridization to an array, or any combination thereof.
- Clonal or subdorsal mutations, CNV's and/or populations of cells may be detected using whole genome sequencing (WGS), whole exome sequencing (WES), single nucleotide polymorphism (SNP) analysis, deep sequencing, targeted gene sequencing, hybridization to an array, or any combination thereof
- WGS whole genome sequencing
- WES whole exome sequencing
- SNP single nucleotide polymorphism
- the gene mutation may be a missense mutation, frameshift indel, inframe indel, splice site mutation, or nonsense mutation.
- Figure 1 The landscape of putative driver gene mutations and recurrent somatic copy number variations in CLL. Somatic mutation information is shown across the 55 putative driver genes and recurrent sCNVs (rows) for 538 primary patient samples (from CLL8 [green], Spanish ICGC [red], DFCI/Broad [blue]) that underwent WES (columns). Blue labels- recurrent sCNVs; Bold labels- putative CLL cancer genes previously identified in Landau et al. 3 ); asterisked labels- additional cancer genes identified in this study. Samples were annotated for IGHV status (black-mutated, white-unmutated; red-unknown), and for exposure to therapy prior to sampling (black-prior therapy; white - no prior therapy; red-unknown prior treatment status).
- Figure 2 Selected novel putative driver gene maps. Individual gene mutation maps for select putative drivers, showing mutation subtype (e.g., missense), position and evidence of mutational hotspots, based on COSMIC database information (remaining gene maps shown in Extended Data Fig. 4).
- FIG. 3A-3B Inferred evolutionary history of CLL.
- A. The proportion in which a recurrent driver is found as clonal or subclonal across the 538 samples is provided (top), along with the individual cancer cell fraction (CCF) values for each sample affected by a driver (tested for each driver with a Fisher's exact test, comparing to the cumulative proportions of clonal and subclonal drivers excluding the driver evaluated). Median CCF values are shown (bottom, bars represent the median and IQR for each driver),
- B. Temporally direct edges are drawn when two drivers are found in the same sample, one in clonal and the other in subclonal frequency.
- edges are used to infer the temporal sequences in CLL evolution, leading from early, through intermediate to late drivers. Note that only driver pairs with at least 5 connecting edges were tested for statistical significance and only drivers connected by at least one statistically significant edge are displayed (see Methods, and Supplementary Table 7).
- FIG. 4A-4B Associations of CLL drivers with cli ical outcome.
- A Kaplan- Meier analysis (with logrank values) for putative drivers with associated impact on progression free survival (PFS) or overall survival (OS) in the cohort of 278 patients that were treated as part of the CLLS trial.
- PFS progression free survival
- OS overall survival
- B Presence of a subclonal driver is associated with shorter PFS, in both the FC and FCR arms, and a trend towards shorter OS.
- FIG. 5A-5C Matched pre-treatment and relapse samples reveal patterns of clo al evolution in relation to therapy, A. The number and proportion of the patterns of clonal evolution of CLLs studied at pre-treatment and at relapse.
- B Selected plots of 2D clustering of pre-treatment and relapse cancer cell fraction (CCF) demonstrating clonal stability of tri(12) (CLL case: GCLL115), concordant increase in CCFs of TP53 and del(ll ⁇ ) (GCLL27), clonal shifts in ATM sSNVs in a sample with clonally stable monoallelic deletion of A TM (GCLL307).
- CCF cancer cell fraction
- Extended Data Figure I Candidate CLL cancer genes discovered in the combined cohort of 538 primary CLL samples. Significantly mutated genes identified in 538 primary CLL. Top panel: the rate of coding mutations (mutations per megabase) per sample. Center panel: Detection of individual gene found to be mutated (sSNVs or sINDELs) in each of the 538 patient samples (columns), color-coded by type of mutation. Only one mutation per gene is shown if multiple mutations from the same gene were found in a sample. Right panel: Q- values (red: Q ⁇ 0.1; purple dashed: Q ⁇ 0.05) and Hugo Symbol gene identification.
- New candidate CLL genes are marked with asterisks (*)
- Left panel The percentages of samples affected with mutations (sSNVs and sINDELs) in each gene.
- Bottom panel plots showing allelic fractions and the spectrum of mutations (sSNVs and sINDELs) for each sample.
- Extended Data Figure 2 Cellular networks and processes affected by putative CLL drivers.
- Putative CLL cancer genes cluster in pathways that are central to CLL biology such as Notch signaling, inflammatory response and B cell receptor signaling.
- proteins that participate in central cellular processes such as DNA damage repair, chromatin modification and mRNA processing, export and translation are also recurrently affected. Boxed in yellow— new CLL subpathways highlighted by the current driver discovery effort. Red circles- putative driver genes previously identified 3 , purple circles- newly identified in the current study,
- Extended Data Figure 3A-3B RNAseq expression data for candidate CLL genes and targeted candidate driver validation.
- Applicants subsequently counted the number of instances in which a mutation was detected ⁇ 'detected') and compared it to the number of instances in which mutation detection had >90% power based on the allelic fraction in the WES and the read depth in the RNAseq data powered'). Overall, Applicants detected 78.1% of putative CLL gene mutations at sites that had >90% power for detection in RNAseq data B.
- Targeted orthogonal validation accession Analysis, Fluidigm
- Extended Data Figure 4A-4V Gene mutation maps for candidate CLL genes. Individual gene mutation maps are shown for all newly identified candidate CLL cancer genes not included in Fig. 2, The plots show mutation subtype (e.g., missense, nonsense etc) and position along the gene.
- Extended Data Figure 5 CLL copy number profiles. Copy number profile across 538 CLLs detected from WES data from primary samples (see Methods).
- Extended Data Figure 6A-6D Annotation of drivers based on clinical characteristics and co-occurrence patterns.
- B Putative drivers affecting greater than 10 patients were assessed for enrichment in samples that received therapy prior to sampling (Fisher's exact test). Putative drivers affecting greater than 10 patients were tested for co-occurrence. Significantly high (C) or low (D) co-occurrences are shown (Q ⁇ 0. ⁇ , Fisher's exact test with BH FDR, after accounting for pri or therapy and IGHV mutation status, see Methods).
- Extended Data Figure 7 Mutation spectrum analysis, clonal vs. subclonal sSNVs, The spectrum of mutation is shown for the clonal and subclonal subsets of coding somatic sSNVs across WES of 538 samples. The rate is calculated by dividing the number of trinucleotides with the specified sSNVs by the covered territory containing the specified trinucleotide. Both clonal and subclonal sSNVs were similarly dominated by C>T transitions at C*pG sites.
- this mutational process that was previously associated with aging 39 , not only predates oncogenic transformation (since clonal mutations will be highly enriched in mutations that precede the malignant transformation 40 ), but also is the dominant mechanism of malignant diversification after transformation in CLL.
- Extended Data Figure 8 The CLL driver landscape in the CLL8 cohort. Somatic mutation information shown across the 55 candidate CLL cancer genes and recurrent sCNVs (rows) for 278 CLL samples collected from patients enrolled on the CLLS clinical trial primary that underwent WES (columns). Recurrent sCNA labels are listed in blue, and candidate CLL cancer genes are listed in bold if previously identified in Landau et & ⁇ . and with an asterisk (*) if newly identified in the current study.
- Extended Data Figure 9 CLLS patient cohort clinical outcome (from 278 patients) information by CLL cancer gene. Kaplan-Meier analysis (with logrank P values) for putative drivers not associated with significant impact on progression free survival (PFS) or overall survival (OS) in the cohort of 278 patients that were treated as part of the CLLS trial . For candidate CLL genes tested here for the first time regarding impact on outcome, a Bonferroni P value is also shown.
- Extended Data Figure 10 Comparison of pre-treatment and relapse cancer cell fraction (CCF) for non-silent mutations in candidate CLL genes across 59 CLLs.
- CCF cancer cell fraction
- CCF increases (red), decreases (blue) or stable (grey) over time are shown (in addition to CLL genes shown in Figure 6).
- a significant change in CCF over time (red or blue) was determined if the 95%CI of the CCF in the pre-treatment and relapse samples did not overlap.
- the term "therapeutic effect” refers to some extent of relief of one or more of the symptoms of a disorder (e.g., a neoplasia or tumor) or its associated pathology.
- “Therapeutically effective amount” as used herein refers to an amount of an agent which is effective, upon single or multiple dose administration to the cell or subject, in prolonging the survivability of the patient with such a disorder, reducing one or more signs or symptoms of the disorder, preventing or delaying, and the like beyond that expected in the absence of such treatment.
- “Therapeutically effective amount” is intended to qualify the amount required to achieve a therapeutic effect.
- a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the "therapeutically effective amount" (e.g., ED50) of the pharmaceutical composition required.
- the physician or veterinarian could start doses of the compounds of the invention employed in a pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
- clinical aggressiveness refers to the average aggressiveness of a cancer. Increased clinical aggressiveness refers to a cancer that is more aggressive as compared to the average.
- a "CLL driver” is any mutation, chromosomal abnormality, or altered gene expression, that contributes to the etiology, progression, severity, aggressiveness, or prognosis of CLL.
- a CLL driver is a mutation that provides a selectable fitness advantage to a CLL cell and facilitates its clonal expansion in the population.
- CLL driver may be used interchangeably with CLL driver event and CLL driver mutation.
- CLL driver mutations occur in genes, genetic loci, or chromosomal regions which may be referred to herein interchangeably as CLL risk alleles, CLL alleles, CLL risk genes, CLL genes, CLL-associated genes and the like.
- staging of chronic lymphocytic leukemia is used to determine treatment of a subject in need thereof.
- Any CLL staging system may be used.
- the Rai staging system and the Binet classification are exemplary systems that may be used in the present invention (Rai KR, et al.: Clinical staging of chronic lymphocytic leukemia. Blood 46 (2): 219-34, 1975; Binet JL, et al. : A new prognostic classification of chronic lymphocytic leukemia derived from a multivariate survival analysis. Cancer 48 (1): 198-206, 1981).
- NCI National Cancer institute
- the Binet classification integrates the number of nodal groups involved with the disease with bone marrow failure. Its major benefit derives from the recognition of a predominantly splenic form of the disease, which may have a better prognosis than in the Rai staging, and from recognition that the presence of anemia or thrombocytopenia has a similar prognosis and does not merit a separate stage. Neither system separates immune from nonimmune causes of cytopenia. Patients with thrombocytopenia or anemia or both, which is caused by extensive marrow infiltration and impaired production (Rai i ii/iV. Binet C) have a poorer prognosis than patients with immune cytopenias.
- Treatments or therapeutic agents contemplated by the present disclosure include but are not limited to immunotherapy, chemotherapy, bone marrow and stem cell transplantation, and others known in the art.
- the treatment described herein is based upon current treatments and clinical trials of treatments used to treat a patient in need thereof suffering from CLL. Doses and administration of treatments may be any regimen used. The treatment may be based on age, health, weight, sex, or ethnicity. The standard of care for the most common cancers can be found on the website of National Cancer Institute (www.cancer.gov/cancertopics). Alternative treatments of leukemia can be found at www.cancer.gov/types/leukemia/hp.
- Standard of care is the current treatment that is accepted by medical experts as a proper treatment for a certain type of disease and that is widely used by healthcare professionals. Standard or care is also called best practice, standard medical care, and standard therapy. Standards of Care for cancer generally include surgery, lymph node removal, radiation, chemotherapy, targeted therapies, antibodies targeting the tumor, and immunotherapy. Immunotherapy can include checkpoint blockers (CBP), chimeric antigen receptors (CARs), and adoptive T-cell therapy.
- CBP checkpoint blockers
- CARs chimeric antigen receptors
- patients diagnosed with stage 0, I, II, III, and IV Chronic Lymphocytic Leukemia are treated with the current standard-of-care first-line treatment.
- This treatment may be a combination of fludarabine (F), cyclophosphamide (C) and rituximab (R).
- alternative treatment options tested in clinical trials are used.
- an alternative treatment of stage 0 CLL is any treatment described herein, not limited to standard treatments for stage 0 CLL.
- the present invention provides for novel stratification of patients suffering from CLL and the stratification may indicate early treatment or observation. Early treatment may be before the appearance of symptoms.
- the present invention may provide for delaying treatment of patients determined to have CLL. Not being bound by a theory, the absence of cancer drivers may indicate a less aggressive CLL.
- the methods of the present invention indicate a subject is treated with a specific treatment.
- the treatment may be different than current treatments.
- the treatments may be the same as current treatments, but administered to a specific population identified with the methods of the present invention.
- patients having different mutations or alterations may have a different response to a specific treatment.
- the identification of novel cancer drivers or alterations allows a more informed treatment choice.
- incorporation of the methods of the present invention allows for delaying treatment or providing earlier treatment as compared to the previous standard of care.
- the identification of a novel cancer driver or alteration may indicate increased aggressiveness of a cancer.
- the subject may have stage 0 CLL or have no symptoms, but have a mutation or alteration of the present invention, such that treatment is indicated. Early treatment of the cancer may lead to longer progression free survival and overall survival.
- the treatment may be any treatment regimen approved or in clinical trials.
- CLL chronic lymphocytic leukemia
- the driver mutations of the present invention are used to determine if a patient should be treated. Not being bound by a theory, monitoring of driver mutation presence can predict spontaneous regressions.
- the methods of the present invention allow an alternative treatment to the current standard of care for Stage 0 CLL.
- treatment for patients grouped as Stage 0 is no treatment.
- the French Cooperative Group on CLL randomly assigned 1,535 patients with previously untreated stage A disease to receive either chlorambucil or no immediate treatment and found no survival advantage for immediate treatment with chlorambucil. (Dighiero G, et al., N Engl J Med 338 (21): 1506-14, 1998).
- a meta-analysis of six trials of immediate versus deferred therapy with chlorambucil showed no difference in overall survival at 10 years. (CLL Trialists' Collaborative Group.
- a therapeutic effect may be achieved using the present invention to stratify patients in the clinical trial based upon the presence of cancer drivers.
- the invention provided herein is useful in determining whether and when to start treatment.
- treatments used to treat Stage I, II, III, and IV CLL are used to treat patients based on identification of the mutations or alterations of the present invention.
- EFS event-free survival
- PFS progression-free survival
- the standard therapy begins when patients develop profound cytopenias, which are consistently indicative of advanced-stage disease, or when they become symptomatic enough that quality of life is substantially impacted, such as with enlarging bulky lymphadenopathy or debilitating symptoms.
- the methods of the present invention allow prediction and early treatment of patients before having advanced-stage disease,
- a patient is treated with a maximal cytoreduction strategy.
- an alternative treatment of a CLL indicated as having an adverse outcome is treated with this strategy.
- This strategy involves using a combination regimen to achieve a durable complete remission.
- FCR is a popular choice for induction therapy (Haliek M, et al. : Lancet 376 (9747): 1164-74, 2010).
- BR bovine rituxirnab
- R-CHOP rituxirnab + cyclophosphamide + doxorubicin + vincristine plus prednisone
- R-CVP eliminating doxorubicin from R-CHOP
- a patient is treated with an avoidance of alkylators and purine analogues strategy.
- the goal of this strategy is to minimize the possibility that subclones with deleterious mutations, which are subsequently resistant to further therapy, will emerge.
- the presence of subclones having any of the driver mutations of the present invention are treated with this alternative strategy.
- This strategy also avoids prolonged cytopenias and the recurrent, long-lasting, sometimes fatal, infections seen after therapy with these agents.
- Initial therapy with a monoclonal antibody, such as rituxirnab (in high doses or more frequent scheduling) with or without maintenance rituxirnab, is consistent with this strategy.
- a patient is treated with a risk-adapted therapy.
- prognostic factors such as the mutational immunoglobulin variable region heavy chain (IgVH), 17p ⁇ , or HQ-status, were used to help inform the choice of therapy.
- Patients with a better prognosis might receive more gentle therapy with fewer short-term and long-term side effects. This approach might also be applicable for older patients with multiple comorbidities.
- Patients with a worse prognosis might receive combination regimens that are used for maximal cytoreduction.
- the methods of the present invention may be used to further determine a prognosis,
- Standard options for CLL treatment are roughly ordered by level of toxic effects, starting with the least toxic options.
- the present invention provides for treating a patient in need thereof based on identification of cancer drivers that indicate an adverse outcome.
- treatment is with a more toxic option.
- the standard options include the following:
- CLL chronic lymphocytic leukemia
- Rituximab is a murine anti-CD20 monoclonal antibody (Mavromatis B, et ai. : J Clin Oncol 21 (9): 1874-81, 2003; O'Brien SM, et al. : J Clin Oncol 19 (8): 2165-70, 2001; Byrd JC, et al. : J Clin Oncol 19 (8): 2153-64, 2001.; Hainsworth ID, et al.: J Clin Oncol 21 (9): 1746-51 , 2003; and Castro JE, et al. : Leukemia 22 (11): 2048-53, 2008), When used alone, higher doses of rituximab or increased frequency or duration of therapy is required for comparable responses to those seen for other indolent lymphomas,
- Ofatumomab is a human anti-CD20 monoclonal antibody (Wierda WG, et al. : J Clin Oncol 28 (10): 1749-55, 2010).
- Obinutuzumab is a human anti-CD20 monoclonal antibody.
- NCT01010061 781 previously untreated patients with coexisting medical problems were randomly assigned to chlorambucil and obinutuzumab versus chlorambucil and rituximab versus chlorambucil alone (Goede V, et al. : N Engl J Med 370 (12): 1101-10, 2014).
- the median PFS was best for the obinutuzumab arm (26.7 months) versus the rituximab arm (16.3 months) versus chlorambucil alone (1 1.1 months ) (hazard ratio [HR], 0.18; 95% confidence interval [CI], 0, 13-0.24; P ⁇ .001) for obinutuzumab and chlorambucil versus chlorambucil alone; for rituximab and chlorambucil versus chlorambucil alone [HR, 0,44; 95% CI, 0.34-0.57; P ⁇ .001 ].
- Idelalisib is an oral inhibitor of the delta isoform of the phosphatidylinositol 3- kinase, which is located in the B-cell receptor-signaling cascade.
- NCT01539512 In a randomized, double-blind, prospective trial (NCT01539512), 220 patients treated mainly with fludarabine-based regimens and who had coexisting medical problems, such as renal dysfunction, received rituximab and idelalisib versus rituximab and placebo (Furman RR, et al: N Engl J Med 370 (11): 997-1007, 2014).
- Ibrutinib is a selective irreversible inhibitor of Bruton tyrosine kinase, a signaling molecule located upstream in the B-eell receptor-signaling cascade. Trials of previously untreated patients and of patients with relapsed or refractory CLL showed durable responses to the oral agent in phase I and II studies (Advani RH, et al.
- Bendamustine is a cytotoxic agent with bifunctional properties of an alkylator and a purine analog (Leoni LM, et al. : Clin Cancer Res 14 (1): 309-17, 2008). In previously treated and untreated patients, bendamustine with rituximab has shown response rates around 70% to 90% (Fischer K, et al. : J Clin Oncol 29 (26): 3559-66, 2011; and lannitto E, et al: Br J Haematol 153 (3): 351-7, 201 1).
- bendamustine showed a better response rate (68% vs. 31%, P ⁇ .0001) and PFS (21 .6 months vs. 8 months) with a median follow-up of 35 months (Knauf WU, et al. : J Clin Oncol 27 (26): 4378- 84, 2009).
- the German CLL Study Group compared bendamustine plus rituximab versus FCR as first-line therapy in patients with CLL who require therapy (Fischer K, et al. : J Clin Oncol 29 (26): 3559-66, 201 1).
- Lenalidomide is an oral immunomodulatory agent with response rates over 50%, with or without rituximab, for patients with previously treated and untreated disease. Prolonged, lower-dose approaches and attention to prevention of tumor lysis syndrome are suggested with this agent (Chen CI, et al. : J Clin Oncol 29 (9): 1175-81, 201 1 ; Chanan-Khan A, et al.: J Clin Oncol 24 (34): 5343-9, 2006, Ferrajoli A, et al : Blood 111 (11): 5291-7, 2008; Strati P, et al.
- o CVP cyclophosphamide plus vincristine plus prednisone
- o CHOP cyclophosphamide plus doxorubicin plus vincristine plus prednisone
- Involved-field radiation therapy Relatively low doses of radiation therapy will affect an excellent response for months or years. Sometimes radiation therapy to one nodal area or the spleen will result in abscopal effect (i.e., the shrinkage of lymph node tumors in untreated sites).
- Alemtuzumab the monoclonal antibody directed at CD52, shows activity in the setting of chemotherapy-resistant disease or high-risk untreated patients with 17p deletion or p53 mutation (Moreton P, et al. : J Clin Oncol 23 (13): 2971 -9, 2005; Parikh SA, et al.: Blood 118 (8): 2062-8, 2011; Pettitt AR, et al. : J Clin Oncol 30 (14): 1647-55, 2012).
- patients are treated if a cancer driver of the present invention is identified.
- the subcutaneous route of delivery for alemtuzumab is preferred to the intravenous route in patients because of the similar efficacy and decreased adverse effects, including less acute allergic reactions that were shown in some nonrandomized reports (Pettitt AR, et al. : J Clin Oncol 30 (14): 1647-55, 2012; Stilgenbauer S, et al. : J Clin Oncol 27 (24): 3994-4001 , 2009; Cortelezzi A, et al. : Leukemia 23 (11): 2027-33, 2009; Osterborg A, et al.: Leukemia 23 (11): 1980-8, 2009, and Gritti G, et al.: Leuk Lymphoma 53 (3): 424-9, 2012).
- Bone marrow and peripheral stem cell transplantations are alternative treatment options (Doney KC, et al.: Bone Marrow Transplant 29 (10): 817-23, 2002; Schetelig J, et al .: J Clin Oncol 21 (14): 2747- 53, 2003; Ritgen M, et al. : Blood 104 (8): 2600-2, 2004; Moreno C, et al.: J Clin Oncol 23 (15): 3433-8, 2005; houri IF, et al.
- Autologous T-cells directed at specific antigen targets were modified by a ientiviral vector to incorporate antigen receptor specificity for the B-cell antigen CD19 and then infused into a previously treated patient (Porter DL, et al. : N Engl J Med 365 (8): 725-33, 2011). A dramatic response lasting 6 months has prompted larger trials of this concept. Ongoing clinical trials are testing the concept of T-cells directed at specific antigen targets with engineered chimeric-antigen receptors (termed CARs (Maus MV, et al. : Blood 123 (17): 2625-35, 2014).
- the present invention provides for treatment with a MEK inhibitor.
- a MEK inhibitor is a chemical or drug that inhibits the mitogen-activated protein kinase kinase enzymes MEK1 and/or MEK2. They can be used to affect the MAPK/ERK pathway which is often overactive in some cancers.
- the present invention provides for identification of CLL driver mutations indicating that treatment with MEK inhibitors should be performed.
- MEK inhibitors may be Trametinib (GSK1 120212), Selumetinib (Janne, Pasi A, et al. (2013).
- the present invention provides for novel targets for therapeutic intervention.
- Cancer drivers may be targeted with any agent that modulates the activity or expression of the target.
- the agent may be an antibody, a soluble polypeptide, a polypeptide agent, a peptide agent, a nucleic acid agent, a nucleic acid ligand, or a small molecule agent.
- the agent may target any of the genes or CNV's described herein.
- the nucleic acid agent may encode for RNAi, a Zinc finger nuclease (ZFN), a Transcription Activator-Like Effector Nuclease (TALEN), or a CRISPR/Cas system.
- the present invention advantageously utilized large sequencing datasets of clinically informative samples to enable the discovery of novel cancer drivers and the network of relationships between the driver events and their impact on disease relapse and clinical outcome.
- the present advantageously allows improved clinical outcomes for patients suffering with
- the CLL8 cohort with two previously reported and non-overlapping WES cohorts 1 '', thereby increasing the size of the cohort to 538 CLLs.
- This cohort size is expected to saturate candidate CLL gene discovery for genes mutated in 5% of patients, and provides 94% and 61% power to detect genes mutated in 3% and 2% of patients, respectively 5 .
- Applicants detected 44 putative CLL. driver genes including 18 CLL mutated drivers that Applicants previously identified 3 , as well as 26 additional putative CLL genes ⁇ Fig. 1-2, Extended Data Fig. 1-2). In total, 33.5% of CLLs harbored mutation in at least one of these 26 additional genes.
- Targeted DNA sequencing as well as variant allele expression by RNAseq demonstrated high rates of orthogonal validation (Extended Data Fig. 3),
- CLL 19 In addition to highlighting novel cellular processes and pathways affected in CLL, many of the 26 additional CLL genes more densely annotated pathways or functional categories previously identified in CLL 19 , including R A processing and export (FUBPI, XP04, ESWR1, NXFI), DNA damage (CHEK2, BRCC3, ELF4 20 and DYRK1A 21 ), chromatin modification (ASXLI, HIST1H IB, BAZ2B, FKZF3) and B cell activity related pathways (TRAF2, TRAF3, CARD11).
- R A processing and export FUBPI, XP04, ESWR1, NXFI
- DNA damage CHEK2, BRCC3, ELF4 20 and DYRK1A 21
- ASXLI chromatin modification
- HIST1H IB HIST1H IB
- BAZ2B BAZ2B
- FKZF3 B cell activity related pathways
- This component of the S40 ribosomal sub unit has not been extensively studied in cancer, although rare mutations have been identified in Diamond- Blackfan anemia 22 .
- CCF cancer-cell fraction
- driver events recurrent sCNVs and candidate CLL gene non-silent mutations
- classifying the driver events based on the relative enrichment of out-degrees vs. in- degrees as early (O ⁇ 0.2 and number of out-degrees > in-degrees). late ⁇ Q ⁇ Q,2 and number of out-degrees ⁇ in-degrees) and intermediate or not powered (£>>0.2. Inter./not powered).
- Out- degrees are defined as instances in which the driver event is clonal and found in the same CLL with another driver event that is subclonal.
- in-degrees are defined as instances in which the driver event is subclonal and found in the same CLL with another driver event that is clonal.
- These tables includes all pairs of driver events (dl, d2) that had at least 5 cases in which the two drivers were detected in the same CLL sample, but one of the drivers is clonal and the other is subclonal.
- a two-tailed binomial test is performed to test whether the pairings are found to be in one order more frequently than the other (i.e., dl c!2 > d2 dl ).
- a multi-hypothesis correction is then applied and the table lists all hypotheses tested.
- An important benefit of the larger cohort size is the enhanced ability to explore relationships between driver lesions based on patterns of their co-occurrence. Focusing on temporal patterns of driver acquisition - based on the distinction between clonal versus subclonal alterations in a cross-sectional analysis - Applicants derived a temporal map for the evolutionary history of CLL. In the context of relapse after first-line fludarabine based therapy, Applicants note highly frequent clonal evolution, and that the future evolutionary trajectories were already anticipated in the pre-treatment sample in one third of cases with WES.
- Immuno-magnetic tumor cell enrichment via CD 19 was performed on all baseline pretreatment samples (Midi MACS, Miltenyi Biotec, Bergisch Gladbach, Germany) achieving a separation of PBMCs into a CD19- positive tumor sample and a CD19-negative normal sample with a purity of >95% by flow cytometry.
- the source of matched normal tissue was PBMC collected following chemoimmunotherapy when the samples were evaluated by flow cytometry as minimal residual disease-negative (Kiel Laboratory, Germany). Samples were used fresh or cryopreserved (with FBS with 10% DMSO) and stored in vapor-phase liquid nitrogen until the time of analysis.
- DNA quality control Applicants used standard Broad Institute protocols as previously described 5,6 . Tumor and normal DNA concentration were measured using PicoGreen® dsDNA Quantitation Reagent (Invitrogen, Carlsbad, CA). A minimum DNA concentration of 5 ng/ ⁇ was required for sequencing. All Illumina sequencing libraries were created with the native DNA. The identities of ail tumor and normal DNA samples were confirmed by mass spectrometric fingerprint genotyping of 95 common SNPs by Fluidigm Genotyping (Fluidigm, San Francisco, CA).
- Standard quality control metrics including error rates, percentage-passing filter reads, and total Gb produced, were used to characterize process performance before downstream analysis. Note that due to a change in the Agilent capture bait set, the NOTCH 1 hotspot was not covered in samples GCLL-199 through GCLL-313, which included 10 samples with NOTCH! c.7544_7545delCT deletions by Sanger sequencing 8 . CLL samples found to harbor NOTCH! mutations by Sanger sequencing were subsequently submitted to targeted sequencing with Illumina TruSeq Custom Amplicon library, and sequenced on an Illumina MiSeq with a mean coverage depth of 1332X.
- samples with NOTCH 1 mutations by Sanger sequencing were also submitted to targeted sequencing.
- samples from patients included in the CLL8 clinical trial Applicants have analyzed 157 WES samples from the cohort Applicants have previously published 5 '. The sequencing reads were realigned to hgl9 and all downstream analysis was done using the same methods as with the sequencing data for the CLL8 cohort.
- Somatic single nucleotide variations were detected using MuTect (Firehose version vl3112); somatic small insertions and deletions were detected using an improved version (manuscript in preparation, Cibulskis et al.,) of Indelocator J .
- the primary improvement is implementation of local reassembly, which results in more accurate allele fraction estimation.
- Applicants filter sSNVs and sINDELs by removing events seen in sequencing data of a large panel of normal samples. Overall, this filtering removed -35% of all candidate somatic mutations, mostly ones with very low allelic fraction.
- MutSig2CV16 to detect candidate cancer genes using three signals of positive selection: (i) increased mutation burden as compared to a background model; (ii) clustering of mutations along the gene; and (iii) enrichment of mutations at likely functional sites.
- Genome-wide copy number analysis Genome-wide copy number profiles of the CLL samples and their patient-matched germline DNA were estimated directly from the WES data, based on the ratio of CLL sample read-depth to the average read-depth observed in normal samples for that region. Applicants observed a high level of agreement between sCNV detection by exome and standard FISH cytogenetics, with the exception of smaller deletions in the region of chromosome 13ql 4, where 14.5% of cases were missed by WES ⁇ Supplementary Table 5). Allelic copy number analysis was then performed by examination of alternate and reference read counts at heterozygous SNP positions (as determined by analysis of the matched normal sample).
- RNA sequencing and data analysis RNA sequencing (RNAseq) was performed as previously described 24 .
- previously published RNAseq data for additional CLL RNA samples were downloaded with permission from the European Genome-Phenome Archive 25 , and processed in an identical fashion to the in-house produced libraries.
- matching WES and RNAseq data were available for 156 samples including 103 samples collected at the DFCI and 53 samples collected by the ICGC.
- RNAseq BAMs were aligned to the hg!9 genome using the TopHat suite. Each somatic base substitution detected by WES was compared to reads at the same location in RNAseq.
- CN T is the local copy number in the tumor cell and CN N is the local copy number in the normal cells (2 in the autosome, and 1 or 2 on the X chromosome depending on gender).
- the allele fraction probability density in the tumor is estimated from the binomial probability density "binopdf over the range of reference allele counts in the tumor "t ref between 0 and t_ref_count:
- AF T is the full range of allele fractions from 0 to 1 and Z is set such that p(AFY) is normalized to 1.
- ⁇ ( ⁇ 1 7 ⁇ ) no longer contains the normal cell component, which simplifies the remaining steps to estimate the mutation cancer cell fraction (CCF).
- CCF mutation cancer cell fraction
- the CCF estimate is integrated over all possible mutation multiplicities "m", the number of mutations per tumor cell.
- the multiplicity m can range from 1 to q hat I or q hat2 (the local somatic absolute copy numbers of each homologous allele in the tumor). Applicants assume that the mutation occurred before or after any local copy number change and each possible multiplicity is given equal weight w m such that the w's are normalized to 1.
- Clustering analysis of sSNVs and sINDELs ie 59 CLL sample pairs Applicants performed WES on matched samples collected at the time of first progression following therapy from 59 of 278 CLL8 subjects (Supplementary Table 9). The median time to progression was 35.1 months (range 5.9-75.5), with relapse samples collected at a median of 7.6 months following documented progression, ail before receipt of subsequent therapy. The two time point CCF clustering procedure was performed as previously described 9 . Clonal evolution between pretreatment and relapse samples was defined based on the presence of mutations with a P ⁇ CCF > 0.1)>0.5. Branched evolution was classified when a dominant clone in the pre-treatment sample was replaced by sibling dominant clone.
- Targeted deep sequencing was performed using microfluidic PGR (Access Array System, Fluidigm). Six unmatched saliva samples were included in this analysis to assist with the quantification of background sequencing error noise. Target-specific primers were designed to flank sites of interest and produce amplicons of 200 bp ⁇ 20 bp. Per well, molecularly barcoded, lilumina-compatible specific oligonucleotides containing sequences complementary to the primer tails were added to the Fluidigm Access Array chip together with genomic DNA samples (20-50 ng of input) such that all amplicons for a given DNA sample shared the same index, and PGR was performed according to the manufacturer's instructions.
- Time to event data were estimated by the Kaplan-Meier method, and differences between groups were assessed using two-sided non- stratified log-rank tests. Additionally, hazard ratios (HR) and 95% confidence intervals (CI) were calculated using unadjusted and adjusted Cox regression modeling. Independent factors for PFS and OS were identified by multivariable analysis using Cox proportional hazards regression models.
- Trisomy 19 Seilmann, L. et al. Trisomy 19 is associated with trisomy 12 and mutated IGHV genes in B-chronic lymphocytic leukaemia. Br J Haematol 138, 217-220, doi :BJH6636 [pii] 10.11 1 1/j .1365-2141.2007.06636.x (2007).
Landscapes
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Engineering & Computer Science (AREA)
- Immunology (AREA)
- Pathology (AREA)
- Analytical Chemistry (AREA)
- Zoology (AREA)
- Genetics & Genomics (AREA)
- Wood Science & Technology (AREA)
- Physics & Mathematics (AREA)
- Biotechnology (AREA)
- Microbiology (AREA)
- Molecular Biology (AREA)
- Hospice & Palliative Care (AREA)
- Biophysics (AREA)
- Oncology (AREA)
- Biochemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
Applicants identified (44) recurrently mutated genes and (11) recurrent somatic copy number variations (CNV) through whole-exome sequencing of 538 chronic lymphocytic leukemia (CLL) and matched germline DNA samples, 278 of which were collected in a prospective clinical trial. These include previously unrecognized cancer drivers (RPS15, IKZF3) and collectively identify RNA processing and export, MYC activity and MAPK signaling as central pathways involved in CLL. Clonality analysis of this large dataset further enabled reconstruction of temporal relationships between driver events. Direct comparison between matched pretreatment and relapse samples from 59 patients demonstrated highly frequent clonal evolution. The discovery of novel cancer genes and the network of relationships between the driver events and their impact on disease relapse and clinical outcome provide novel treatment options.
Description
METHODS AND COMPOSITIONS FOR USE OF DRIVER MUTATIONS IN CLL
RELATED APPLICATIONS AND INCORPORATION BY REFERENCE
[0001] This application claims priority and benefit of U.S. provisional application serial number 62/244,533, filed October 21, 2015.
[0002] Reference is made to U.S. provisional patent application Serial No. 62/,053,697 filed September 22, 2014 and U.S. provisional patent application Serial No. 62/181,715, filed June 18, 2015. Reference is also made to International patent application serial numbers PCT/US2012/068633, filed December 7, 2012 and PCT/US2015/051340, filed September 22, 2015.
[0003] The foregoing applications, and all documents cited therein or during their prosecution ("appln cited documents") and all documents cited or referenced in the appln cited documents, and all documents cited or referenced herein ("herein cited documents"), and all documents cited or referenced in herein cited documents, together with any manufacturer's instructions, descriptions, product specifications, and product sheets for any products mentioned herein or in any document incorporated by reference herein, are hereby incorporated herein by reference, and may be employed in the practice of the invention. More specifically, all referenced documents are incorporated by reference to the same extent as if each individual document was specifically and individually indicated to be incorporated by reference.
FEDERAL FUNDING LEGEND
[0004] This invention was made with government support under grant numbers CA182461 , CA184922, and CA180861 awarded by the National Institutes of Health. The government has certain rights in the invention.
FIELD OF THE INVENTION
[0005] The present invention provides methods for treatment and prognosis of chronic lymphocytic leukemia (CLL) using novel cancer driver mutations.
B ACKGROUND OF THE INVENTION
[0006] In recent years, unbiased massively parallel sequencing of whole exomes (WES) in chronic lymphocytic leukemia (CLL) has yielded fresh insights into the genetic basis of this disease1"4. Two important constraints have limited previous WES analyses. First, cohort size is
critical for statistical inference of cancer drivers5, and prior CLL WES series3 had a power of only 68%, 23% and 7%, to detect putative CLL genes mutated in 5%, 3% and 2% of patients, respectively (www.tumorportal.org/power)5. Limited cohort size has also curtailed the ability to effectively learn the relationships between CLL driver events, such as their co-occurrence and the temporal order of their acquisition. Second, the composition of the cohort of prior WES studies has limited the ability to accurately determine the impact of drivers and clonal heterogeneity on clinical outcome since they included samples collected at variable times from subjects exposed to a variety of therapies. Understanding the genetic alterations that drive tumorigenesis and how they evolve over the course of disease and therapy are central questions in cancer biology. Thus, identifying novel cancer drivers is necessary to improve treatment and diagnosis of CLL.
[0007] Citation or identification of any document in this application is not an admission that such document is available as prior art to the present invention.
SUMMARY OF THE INVENTION
[0008] It is an objective of the present invention to provide novel cancer driver alterations for use in improved treatment of patients suffering from CLL. Applicants identified 44 recurrently mutated genes and 11 recurrent somatic copy number variations through whole-e ome sequencing of 538 chronic lymphocytic leukemia (CLL) and matched germline DNA samples, 278 of which were collected in a prospective clinical trial. These include previously unrecognized cancer drivers (RPS15, IKZF3) and collectively identify RNA processing and export, MYC activity and MAPK signaling as central pathways involved in CLL. Clonality analysis of this large dataset further enabled reconstruction of temporal relationships between driver events. Direct comparison between matched pre-treatment and relapse samples from 59 patients demonstrated highly frequent clonal evolution.
[0009] Specifically, Applicants analyzed WES data from 538 CLLs, including 278 pre- treatment samples collected from subjects enrolled on the phase III CLL8 study13. This trial established the combination of fludarabine (F), cyclophosphamide (C) and rituximab (R) as the current standard-of-care first-line treatment for patients of good physical fitness, with a median of >6 years of follow-up. Applicants discovered novel CLL cancer genes, performed a
comprehensive genetic characterization of samples from patients prior to exposure to a uniform and contemporary treatment, and uncovered features contributing to relapse from this therapy.
[0010] In a first aspect the present invention provides for a method of treating chronic lymphocytic leukemia (CLL) in a subject in need thereof comprising identifying the presence of gene mutations and somatic copy number variations (CNV) in a genomic DNA sample obtained from the subject, wherein the mutated genes and somatic copy number variations (CNV) comprise del(13q), SF3B1, A TM, del(llq), tril2, TP53, NOTCH 1, POT1, amp(2p), del(17p), CHD2, XPOl, RPS15, BIRC3, BRAF, IGLL5, d l(8p), MGA, MYD88, del(6q21), EGR2, FBXW7, ampfSq), DDX3X, KRAS, BCOR, IKZF3, MAP2K1, ZMYM3, IRF4, SAMHD1, BAZ2A, CARD! I, FUBP1, HIST1H1E, MED12, NRAS, NXF1, del (18p), DYRK1A, PTPNll, del(20p), ELF 4, TRAF2, XP04, tril9, BRCC3, Erf SRI, FAM50A, TRAF3, ASXL!, CHEK2, GNB1, HISTIHIB, and PIMI, wherein the subject is treated if the presence of a mutated gene and/or somatic copy number variation (CNV) is identified. The patient may have stage 0, 1, II, III, or IV CLL. Preferably, the method is performed before a patient has symptoms. The method may be performed before treatment, during treatment, or after treatment. The method may be performed when the patient is in remission. The presence of mutations in NRAS, KRAS, BRAF, and/or MAP2K1 may indicate treating with an inhibitor of the MAPK-ERK pathway. The mutation in BRjiF may be a mutation that is not the canonical V600E mutation. In this instance the treatment may include a MEK inhibitor and would not indicate the use of a BRAF inhibitor. The presence of mutations in RPS15 may indicate an adverse outcome and the subject is treated with an alternative treatment regimen. The presence of mutations in IKZF3 may indicate an adverse outcome and the subject is treated with an alternative treatment regimen. The mutation in IKZF3 may be an L162R substitution. The presence of mutations in delflSq), MYD88, and/ or ( 7/7/2 may indicate decreased clinical aggressiveness. The presence of mutations in TP53, SF3B1 and/or XPOl may indicate shorter progression-free survival (PFS) and overall survival (OS).
[0011] The genomic DNA sample may be obtained prior to treatment by any methods known in the art. Not being bound by a theory, obtaining the sample prior to treatment allows the detection of the mutations or alterations in copy number of the present invention in clonal and subclonal populations of cells present in a cancer cell fraction (CCF) before treatment. Identification of mutations or alterations in copy number can be used to predict resistence to a treatment and efficacy of a treatment.
[0012] The method may further comprising determining mutations in IGHV, wherein the presence of unmutated IGHV indicates increased clinical aggressiveness and the presence of mutated IGHV indicates decreased clinical aggressiveness.
[0013] In another aspect, the present invention provides for a method of determining whether a subject having chronic lymphocytic leukemia (CLL) would derive a clinical benefit of early treatment comprising determining the presence of gene mutations and somatic copy number variations (CNV) in a genomic DNA sample obtained from the subject, wherein the mutated genes and somatic copy number variations (CNV) comprise del(13q), SF3B1, ATM, del(llq), trill, TPS 3, NOTCH 1, POT1, amp(2p), del(17p), CHD2, XPOl, RPS15, BIRC3, BRAF, IGLL5, del(8p), MGA, MYD88, del(6q21), EGR2, FBXW7, amp(8q), DDX3X, KRAS, BCOIi IKZF3, MAP2K1, 7MYM3, IRF4, SAMHD!, BAZ2A, CARDIl, FUBP1, HISTIH!E, MED 12, NRAS, NXFI, del (18p), DYRK1A, PTPNll, delQOp), ELF 4, TRAF2, XP04, in 19. BRCC3, EWSR1, FAM50A, TRAF3, ASXL1, CHEK2, GNBl, HISTIHIB, and P!Ml, wherein the presence of a mutated gene and/or somatic copy number variation (CNV) indicates that the subject would derive a clinical benefit of early treatment,
[0014] In another aspect, the present invention provides for a method of predicting survivability of a subject having chronic lymphocytic leukemia (CLL) comprising determining the presence of gene mutations and somatic copy number variations (CN V) in a genomic DN A sample obtained from the subject, wherein the mutated genes and somatic copy number variations (CNV) co pnsQdeI(I3q), SF3B1, ATM, del(l lq), trill, TP53, NOTCH!, POT1, amp(2p), del(17p), CIID2, XPOl, RPS15, BIRC3, BRAF, IGLL5, del(8p), MGA, MYD88, del(6q21), EGR2, FBXW7, amp(8q), DDX3X, KRAS, BCOR, IKZF3, MAP2K1, 7MYM3, IRF4, SAMHD!, BAZ2A, CARDIl, FlJBPl, HISTIHIE, MED12, NRAS, NXFI, del (18p), DYRK1A, PTPNll, del(20p), ELF 4, TRAF2, XP04, tril9, BRCC3, EWSRl, FAM50A, TRAP 3, ASXL1, CHEK2, GNBl, HISTIHIB, and PIM1, wherein the presence of a mutated gene and/or somatic copy number variation (CNV) indicates that the subject is less likely to survive.
[0015] In another aspect, the present invention provides for a method of identifying a candidate subject for a clinical trial for a treatment protocol for chronic lymphocytic leukemia (CLL) comprising determining the presence of gene mutations and somatic copy number variations (CNV) in a genomic DNA sample obtained from the subject, wherein the mutated genes and somatic copy number variations (CNV) comprise del(13q), SF3B1, ATM, del/1 Iq),
trill, TP 53, NOTCH!, POT I, amp(lp), deI(T7p), CHD2, XPOl, RPS15, B1RC3, BRAF, IGLL5, del(8p), MGA, MYD88, deMqll), EGR2, FBXW7, mpfSq), DDX3X, KRAS, BCOR, IKZF3, MAP1K1, ZMYM3, IRF4, SAMHD1, BAZ1A, CARDll, FUBPl, H1ST1H1E, MED 12, NRAS, NXFI, del (18p), DYRKIA, PTPNII, del(20p), ELF 4, TRAF2, XP04, tnl9, BRCC3, EWSRI, FAS 150 1. TRAF3, ASXLl, CHEK2, GNB1, HISTIHIB, and PIML wherein the presence of a mutated gene and/or somatic copy number variation (CNV) indicates that the subject is a candidate for the clinical trial.
[0016] In another aspect, the present invention provides for a method of detecting chronic lymphocytic leukemia (CLL) in a subject comprising determining the presence of gene mutations and somatic copy number variations (CNV) in a genomic DNA sample obtained from the subject, wherein the mutated genes and somatic copy number variations (CNV) comprised?/^), SF3B1, Am, del/1 Iq), tnl2, TP 53, NOTCH 1, POT1, amp(2p), del(17p), CHD2, XPOl, RPSI5, BIRC3, BRAF, IGLL5, del(8p), MGA, MYD88, del(6q2l), EGR2, FBXW7, αηιρβ , DDX3X, KRAS, BCOR, IKZF3, MAP2K1, ZMYM3, IRF4, SAMHD1, BAZ2A, CARDll, FUBPl, HISTIHIE, MED 12, NRAS, NXFI, del (18p), DYRKIA, PTPNII, del (20p), ELF 4, TRAF2, XP04, tril9, BRCC3, EWSRI, FAM50A, TRAF3, ASXI !. CHEK2, GNB1, HISTIHIB, and P!Ml , whereby the presence of one or more mutations and/or CNV indicates that the subject has a greater than 90% probability of having CLL.
[0017] In another aspect, the present invention provides for a method of identifying a subject at elevated risk of having CLL with rapid disease progression comprising: (a) analyzing genomic DNA in a sample obtained from a subject having or suspected of having chronic lymphocytic leukemia (CLL) for the presence of gene mutations and somatic copy number variations (CNV), wherein the mutated genes and somatic copy number variations (CNV) comprise£fe/{73¾), .SV-'.?/>7, A TM, del(llq), tril2, TP53, NOTCH 1, POT I, amp(lp), del(l7p), CHDl, XPOl, RPS15, BIRC3, BRAF, IGLL5, del(8p), MGA, MYD88, deUfiqll), EGRl, FBXW7, amp(8q), DDX3X, KRAS, BCOR, IKZF3, MA 2 h i, ZMYM3, TRF4, SAMHDl, BAZIA, CARDll, FUBPl, HISTIHIE, MED 12. NRAS, NXFI, del (I8p), DYRKIA, PTPNII, del(20p), ELF 4, TRAF2, XP04, tril9, BRCC3, EWSRI, FAM50A, TRAF3, ASXLl, CHEK2, GNBl, HISTIHIB, and PIM1; (b)determining whether the mutations and/or somatic copy number variations are clonal or subclonal, and (c) identifying the subject as a subject at elevated risk of having CLL with rapid disease progression if at least one mutation and/or somatic copy number variation is
subdorsal. The method may further comprise treating a subject for CLL identified as a subject at elevated risk of having CLL with rapid disease progression.
[0018] Any of the methods may include determining mutations in more than one risk allele. Any of the methods may be repeated every month, 2 months, 3 months, 4 months, five months, 6 months 8 months, or one year or at any time where there is a change in clinical status. The genomic DNA sample may be obtained from peripheral blood, bone marrow, or lymph node tissue. The genomic DNA may be analyzed using whole genome sequencing (WGS), whole exome sequencing (WES), single nucleotide polymorphism (SNP) analysis, deep sequencing, targeted gene sequencing, hybridization to an array, or any combination thereof. Clonal or subdorsal mutations, CNV's and/or populations of cells may be detected using whole genome sequencing (WGS), whole exome sequencing (WES), single nucleotide polymorphism (SNP) analysis, deep sequencing, targeted gene sequencing, hybridization to an array, or any combination thereof The gene mutation may be a missense mutation, frameshift indel, inframe indel, splice site mutation, or nonsense mutation.
[0019] Accordingly, it is an object of the invention to not encompass within the invention any previously known product, process of making the product, or method of using the product such that Applicants reserve the right and hereby disclose a disclaimer of any previously known product, process, or method. It is further noted that the invention does not intend to encompass within the scope of the invention any product, process, or making of the product or method of using the product, which does not meet the written description and enablement requirements of the USPTO (35 I. S C. § 112, first paragraph) or the EPO (Article 83 of the EPC), such that Applicants reserve the right and hereby disclose a disclaimer of any previously described product, process of making the product, or method of using the product.
[0020] It is noted that in this disclosure and particularly in the claims and/or paragraphs, terms such as "comprises", "comprised", "comprising" and the like can have the meaning attributed to it in U. S. Patent law; e.g., they can mean "includes", "included", "including", and the like; and that terms such as "consisting essentially of and "consists essentially of have the meaning ascribed to them in U.S. Patent law, e.g., they allow for elements not explicitly recited, but exclude elements that are found in the prior art or that affect a basic or novel characteristic of the invention. Nothing herein is intended as a promise.
[0021] These and other embodiments are disclosed or are obvious from and encompassed by, the following Detailed Description.
BRIEF DESCRIPTION OF THE DRAWINGS
[0022] The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.
[0023] The following detailed description, given by way of example, but not intended to limit the invention solely to the specific embodiments described, may best be understood in conjunction with the accompanying drawings.
[0024] Figure 1 : The landscape of putative driver gene mutations and recurrent somatic copy number variations in CLL. Somatic mutation information is shown across the 55 putative driver genes and recurrent sCNVs (rows) for 538 primary patient samples (from CLL8 [green], Spanish ICGC [red], DFCI/Broad [blue]) that underwent WES (columns). Blue labels- recurrent sCNVs; Bold labels- putative CLL cancer genes previously identified in Landau et al.3); asterisked labels- additional cancer genes identified in this study. Samples were annotated for IGHV status (black-mutated, white-unmutated; red-unknown), and for exposure to therapy prior to sampling (black-prior therapy; white - no prior therapy; red-unknown prior treatment status).
[0025] Figure 2: Selected novel putative driver gene maps. Individual gene mutation maps for select putative drivers, showing mutation subtype (e.g., missense), position and evidence of mutational hotspots, based on COSMIC database information (remaining gene maps shown in Extended Data Fig. 4).
[0026] Figure 3A-3B: Inferred evolutionary history of CLL. A. The proportion in which a recurrent driver is found as clonal or subclonal across the 538 samples is provided (top), along with the individual cancer cell fraction (CCF) values for each sample affected by a driver (tested for each driver with a Fisher's exact test, comparing to the cumulative proportions of clonal and subclonal drivers excluding the driver evaluated). Median CCF values are shown (bottom, bars represent the median and IQR for each driver), B. Temporally direct edges are drawn when two drivers are found in the same sample, one in clonal and the other in subclonal frequency. These edges are used to infer the temporal sequences in CLL evolution, leading from early, through
intermediate to late drivers. Note that only driver pairs with at least 5 connecting edges were tested for statistical significance and only drivers connected by at least one statistically significant edge are displayed (see Methods, and Supplementary Table 7).
[0027] Figure 4A-4B. Associations of CLL drivers with cli ical outcome. A, Kaplan- Meier analysis (with logrank values) for putative drivers with associated impact on progression free survival (PFS) or overall survival (OS) in the cohort of 278 patients that were treated as part of the CLLS trial. For candidate CLL genes tested here for the first time regarding impact on outcome, a Bonferroni Q value is also shown. B. Presence of a subclonal driver is associated with shorter PFS, in both the FC and FCR arms, and a trend towards shorter OS.
[0028] Figure 5A-5C: Matched pre-treatment and relapse samples reveal patterns of clo al evolution in relation to therapy, A. The number and proportion of the patterns of clonal evolution of CLLs studied at pre-treatment and at relapse. B. Selected plots of 2D clustering of pre-treatment and relapse cancer cell fraction (CCF) demonstrating clonal stability of tri(12) (CLL case: GCLL115), concordant increase in CCFs of TP53 and del(ll^) (GCLL27), clonal shifts in ATM sSNVs in a sample with clonally stable monoallelic deletion of A TM (GCLL307). Red coloring was added when greater than half of the CCF probability indicated >0.1 increase in CCF at relapse. C. Clonal evolution of CLL drivers. Left panel - for each driver with at least 4 instances detected across the 59 CLLs, the proportion of instances where the CCF increased (red), decreased (blue) or remained stable (grey) over time is shown (see Methods for details of the statistical analysis). The driver events were distributed to three main groups: predominately stable events (top); predominately increasing CCF (middle), and all other patterns (bottom). Right panel - Comparison (modal CCF with 95%CI) between pre-treatment and relapse samples for select CLL drivers (see Extended Data Fig. 10 for the remaining driver events from the cohort of 59 CLLs).
[0029] Extended Data Figure I : Candidate CLL cancer genes discovered in the combined cohort of 538 primary CLL samples. Significantly mutated genes identified in 538 primary CLL. Top panel: the rate of coding mutations (mutations per megabase) per sample. Center panel: Detection of individual gene found to be mutated (sSNVs or sINDELs) in each of the 538 patient samples (columns), color-coded by type of mutation. Only one mutation per gene is shown if multiple mutations from the same gene were found in a sample. Right panel: Q- values (red: Q<0.1; purple dashed: Q<0.05) and Hugo Symbol gene identification. New
candidate CLL genes are marked with asterisks (*) Left panel: The percentages of samples affected with mutations (sSNVs and sINDELs) in each gene. Bottom panel: plots showing allelic fractions and the spectrum of mutations (sSNVs and sINDELs) for each sample.
[0030] Extended Data Figure 2: Cellular networks and processes affected by putative CLL drivers. Putative CLL cancer genes cluster in pathways that are central to CLL biology such as Notch signaling, inflammatory response and B cell receptor signaling. In addition, proteins that participate in central cellular processes such as DNA damage repair, chromatin modification and mRNA processing, export and translation are also recurrently affected. Boxed in yellow— new CLL subpathways highlighted by the current driver discovery effort. Red circles- putative driver genes previously identified3 , purple circles- newly identified in the current study,
[0031] Extended Data Figure 3A-3B: RNAseq expression data for candidate CLL genes and targeted candidate driver validation. A. Matched RNAseq and WES data were available for 156 CLLs (103 CLLs previously reported in Landau et al.3 and 53 CLLs from the ICGC studies1). From the WES of these 156 cases, Applicants identified 318 driver mutations (sSNVs and sINDELs). For each site, Applicants quantified the number of alternate reads corresponding to the somatic mutation in matched RNAseq data. Applicants subsequently counted the number of instances in which a mutation was detected {'detected') and compared it to the number of instances in which mutation detection had >90% power based on the allelic fraction in the WES and the read depth in the RNAseq data powered'). Overall, Applicants detected 78.1% of putative CLL gene mutations at sites that had >90% power for detection in RNAseq data B. Targeted orthogonal validation (Access Array System, Fluidigm) was performed for 71 mutations (sSNVs and sINDELs) in putative CLL genes, affecting 47 CLLs from the CLLS cohort (selected based on sample availability). With a mean depth of coverage of 7472X, 65 of the 71 mutations (91.55%) validated, with a higher variant allele fraction compared with normal sample DNA (binomial P <0.01).
[0032] Extended Data Figure 4A-4V: Gene mutation maps for candidate CLL genes. Individual gene mutation maps are shown for all newly identified candidate CLL cancer genes not included in Fig. 2, The plots show mutation subtype (e.g., missense, nonsense etc) and position along the gene.
[0033] Extended Data Figure 5: CLL copy number profiles. Copy number profile across 538 CLLs detected from WES data from primary samples (see Methods).
[0034] Extended Data Figure 6A-6D: Annotation of drivers based on clinical characteristics and co-occurrence patterns. A, Putative drivers affecting greater than 10 patients were assessed for enrichment in IGHV mutated vs. unmutated CLL subtype (Fisher's exact test, magenta line denotes P = 0.05). B. Putative drivers affecting greater than 10 patients were assessed for enrichment in samples that received therapy prior to sampling (Fisher's exact test). Putative drivers affecting greater than 10 patients were tested for co-occurrence. Significantly high (C) or low (D) co-occurrences are shown (Q<0. \, Fisher's exact test with BH FDR, after accounting for pri or therapy and IGHV mutation status, see Methods).
[0035] Extended Data Figure 7: Mutation spectrum analysis, clonal vs. subclonal sSNVs, The spectrum of mutation is shown for the clonal and subclonal subsets of coding somatic sSNVs across WES of 538 samples. The rate is calculated by dividing the number of trinucleotides with the specified sSNVs by the covered territory containing the specified trinucleotide. Both clonal and subclonal sSNVs were similarly dominated by C>T transitions at C*pG sites. Thus, this mutational process that was previously associated with aging39, not only predates oncogenic transformation (since clonal mutations will be highly enriched in mutations that precede the malignant transformation40), but also is the dominant mechanism of malignant diversification after transformation in CLL.
[0036] Extended Data Figure 8: The CLL driver landscape in the CLL8 cohort. Somatic mutation information shown across the 55 candidate CLL cancer genes and recurrent sCNVs (rows) for 278 CLL samples collected from patients enrolled on the CLLS clinical trial primary that underwent WES (columns). Recurrent sCNA labels are listed in blue, and candidate CLL cancer genes are listed in bold if previously identified in Landau et &\. and with an asterisk (*) if newly identified in the current study.
[0037] Extended Data Figure 9: CLLS patient cohort clinical outcome (from 278 patients) information by CLL cancer gene. Kaplan-Meier analysis (with logrank P values) for putative drivers not associated with significant impact on progression free survival (PFS) or overall survival (OS) in the cohort of 278 patients that were treated as part of the CLLS trial . For candidate CLL genes tested here for the first time regarding impact on outcome, a Bonferroni P value is also shown.
[0038] Extended Data Figure 10: Comparison of pre-treatment and relapse cancer cell fraction (CCF) for non-silent mutations in candidate CLL genes across 59 CLLs. For each CLL gene mutated across the 59 CLLs that were sampled longitudinally, the modal CCF is compared between the pre-treatment and relapse samples. CCF increases (red), decreases (blue) or stable (grey) over time are shown (in addition to CLL genes shown in Figure 6). A significant change in CCF over time (red or blue) was determined if the 95%CI of the CCF in the pre-treatment and relapse samples did not overlap.
DETAILED DESCRIPTION OF THE INVENTION
[0039] The term "therapeutic effect" refers to some extent of relief of one or more of the symptoms of a disorder (e.g., a neoplasia or tumor) or its associated pathology. "Therapeutically effective amount" as used herein refers to an amount of an agent which is effective, upon single or multiple dose administration to the cell or subject, in prolonging the survivability of the patient with such a disorder, reducing one or more signs or symptoms of the disorder, preventing or delaying, and the like beyond that expected in the absence of such treatment. "Therapeutically effective amount" is intended to qualify the amount required to achieve a therapeutic effect. A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the "therapeutically effective amount" (e.g., ED50) of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in a pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
[0040] The term "clinical aggressiveness" refers to the average aggressiveness of a cancer. Increased clinical aggressiveness refers to a cancer that is more aggressive as compared to the average.
[0041] A "CLL driver" is any mutation, chromosomal abnormality, or altered gene expression, that contributes to the etiology, progression, severity, aggressiveness, or prognosis of CLL. In some aspects, a CLL driver is a mutation that provides a selectable fitness advantage to a CLL cell and facilitates its clonal expansion in the population. CLL driver may be used interchangeably with CLL driver event and CLL driver mutation. CLL driver mutations occur in
genes, genetic loci, or chromosomal regions which may be referred to herein interchangeably as CLL risk alleles, CLL alleles, CLL risk genes, CLL genes, CLL-associated genes and the like.
[0042] In one embodiment, staging of chronic lymphocytic leukemia (CLL) is used to determine treatment of a subject in need thereof. Any CLL staging system may be used. The Rai staging system and the Binet classification are exemplary systems that may be used in the present invention (Rai KR, et al.: Clinical staging of chronic lymphocytic leukemia. Blood 46 (2): 219-34, 1975; Binet JL, et al. : A new prognostic classification of chronic lymphocytic leukemia derived from a multivariate survival analysis. Cancer 48 (1): 198-206, 1981). Additionally, a National Cancer institute (NCI)-sponsored working group has formulated standardized guidelines for criteria related to eligibility, response, and toxic effects to be used in future clinical trials in CLL (Hallek M, et al. : Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines. Blood 1 1 1 (12): 5446-56, 2008).
[0043] The Binet classification integrates the number of nodal groups involved with the disease with bone marrow failure. Its major benefit derives from the recognition of a predominantly splenic form of the disease, which may have a better prognosis than in the Rai staging, and from recognition that the presence of anemia or thrombocytopenia has a similar prognosis and does not merit a separate stage. Neither system separates immune from nonimmune causes of cytopenia. Patients with thrombocytopenia or anemia or both, which is caused by extensive marrow infiltration and impaired production (Rai i ii/iV. Binet C) have a poorer prognosis than patients with immune cytopenias. The International Workshop on CLL has recommended integrating the Rai and Binet systems as follows: A(0), A (I), A(II); B(I), B(II); and C(III), C'i lY) The NCI-sponsored working group has published guidelines for the diagnosis and treatment of CLL in both clinical trial and general practice settings. Use of these systems allows comparison of clinical results and establishment of therapeutic guidelines. In preferred embodiments any of these classification systems may be used with the methods of the present invention.
[0044] Treatments or therapeutic agents contemplated by the present disclosure include but are not limited to immunotherapy, chemotherapy, bone marrow and stem cell transplantation, and others known in the art. In one embodiment, the treatment described herein is based upon current treatments and clinical trials of treatments used to treat a patient in need thereof suffering from CLL. Doses and administration of treatments may be any regimen used. The treatment may be based on age, health, weight, sex, or ethnicity. The standard of care for the most common cancers can be found on the website of National Cancer Institute (www.cancer.gov/cancertopics). Alternative treatments of leukemia can be found at www.cancer.gov/types/leukemia/hp. The standard of care is the current treatment that is accepted by medical experts as a proper treatment for a certain type of disease and that is widely used by healthcare professionals. Standard or care is also called best practice, standard medical care, and standard therapy. Standards of Care for cancer generally include surgery, lymph node removal, radiation, chemotherapy, targeted therapies, antibodies targeting the tumor, and immunotherapy. Immunotherapy can include checkpoint blockers (CBP), chimeric antigen receptors (CARs), and adoptive T-cell therapy.
[0045] In one embodiment, patients diagnosed with stage 0, I, II, III, and IV Chronic Lymphocytic Leukemia are treated with the current standard-of-care first-line treatment. This
treatment may be a combination of fludarabine (F), cyclophosphamide (C) and rituximab (R). In one embodiment, alternative treatment options tested in clinical trials are used. In one embodiment, an alternative treatment of stage 0 CLL is any treatment described herein, not limited to standard treatments for stage 0 CLL. Not being bound by a theory, the present invention provides for novel stratification of patients suffering from CLL and the stratification may indicate early treatment or observation. Early treatment may be before the appearance of symptoms. The present invention may provide for delaying treatment of patients determined to have CLL. Not being bound by a theory, the absence of cancer drivers may indicate a less aggressive CLL.
[0046] In one embodiment, the methods of the present invention indicate a subject is treated with a specific treatment. The treatment may be different than current treatments. The treatments may be the same as current treatments, but administered to a specific population identified with the methods of the present invention. Not being bound by a theory, patients having different mutations or alterations may have a different response to a specific treatment. Not being bound by a theory, the identification of novel cancer drivers or alterations allows a more informed treatment choice. Not being bound by a theory, incorporation of the methods of the present invention allows for delaying treatment or providing earlier treatment as compared to the previous standard of care. Not being bound by a theory the identification of a novel cancer driver or alteration may indicate increased aggressiveness of a cancer. The subject may have stage 0 CLL or have no symptoms, but have a mutation or alteration of the present invention, such that treatment is indicated. Early treatment of the cancer may lead to longer progression free survival and overall survival. The treatment may be any treatment regimen approved or in clinical trials.
[0047] Standard treatments of chronic lymphocytic leukemia (CLL) ranges from periodic observation with treatment of infectious, hemorrhagic, or immunologic complications to a variety of therapeutic options, including steroids, alkylating agents, purine analogs, combination chemotherapy, monoclonal antibodies, and transplant options. CLL is most often treated in a conservative fashion.(Gribben JG, et al . J Clin Oncol 29 (5): 544-50, 2011), In asymptomatic patients, treatment may be deferred until the patient becomes symptomatic as the disease progresses. Since the rate of progression may vary from patient to patient, with long periods of stability and sometimes spontaneous regressions, frequent and careful observation is required to monitor the clinical course (Del Giudice I, et al., Blood 114 (3): 638-46, 2009). The methods of
the present invention may be used to monitor such patients. In one embodiment, the driver mutations of the present invention are used to determine if a patient should be treated. Not being bound by a theory, monitoring of driver mutation presence can predict spontaneous regressions.
[0048] In one embodiment, the methods of the present invention allow an alternative treatment to the current standard of care for Stage 0 CLL. Prior to the present invention, treatment for patients grouped as Stage 0 is no treatment. The French Cooperative Group on CLL randomly assigned 1,535 patients with previously untreated stage A disease to receive either chlorambucil or no immediate treatment and found no survival advantage for immediate treatment with chlorambucil. (Dighiero G, et al., N Engl J Med 338 (21): 1506-14, 1998). A meta-analysis of six trials of immediate versus deferred therapy with chlorambucil (including the aforementioned trial by the French Cooperative Group) showed no difference in overall survival at 10 years. (CLL Trialists' Collaborative Group. J Natl Cancer Inst 91 (10): 861-8, 1999). Not being bound by a theory, a therapeutic effect may be achieved using the present invention to stratify patients in the clinical trial based upon the presence of cancer drivers. The invention provided herein is useful in determining whether and when to start treatment.
[0049] In other embodiments, treatments used to treat Stage I, II, III, and IV CLL are used to treat patients based on identification of the mutations or alterations of the present invention. Several decades of large, randomized, prospective trials of previously untreated patients have demonstrated statistically significant improvements in response rates, event-free survival (EFS), and progression-free survival (PFS) with comparison of combinations of drugs versus single- agent alkyiators, (Clin Adv Hematol Oncol 5 (3 Suppl 5): 1-14; quiz 15-6, 2007; and Montserrat E, et al. Blood 107 (4): 1276-83, 2006), Three trials have shown statistically significant improvement in overall survival (OS) (Rai KR, et al., Blood 14 (22): A-536, 2009; Hallek M, et al., Lancet 376 (9747): 1 164-74, 2010, and Byrd JC, et al., N Engl J Med 371 (3): 213-23, 2014).
[0050] The first trial, a comparison of chlorambucil versus fludarabine for untreated chronic lymphocytic leukemia (CLL), after 15 years of median follow-up, showed improved median OS for patients on the fludarabine regimen at 63 months versus 59 months (P :=: .04), and an improved percentage of patients were alive at 8 years (31% vs. 19%, P = .04) (Rai KR, et al., Blood 114 (22): A-536, 2009, and Rai KR, et al,, N Engl J Med 343 (24): 1750-7, 2000).
[0051] The second trial, which had 817 untreated patients, compared FCR (fludarabine + cyclophosphamide + rituximab) versus FC (fludarabine + cyclophosphamide) with a median
follow-up of 38 months and showed improved OS at 3 years for the rituxirnab combination (i.e., 87% vs. 83%, P = .01 (Haliek M, et al., Lancet 376 (9747): 64-74, 2010).
[0052] A prospective randomized trial of 391 patients with relapsed or refractor}' CLL and small lymphocytic lymphoma (SLL) compared ibrutinib with ofatumumab. With a median follow-up of 9.4 months, the 12-month OS favored ibrutinib (90% to 81%, HR, 0.43; P = .005) (Byrd JC, et al, N Engl J Med 371 (3): 213-23, 2014).
[0053] In the absence of a clinical trial, the standard therapy begins when patients develop profound cytopenias, which are consistently indicative of advanced-stage disease, or when they become symptomatic enough that quality of life is substantially impacted, such as with enlarging bulky lymphadenopathy or debilitating symptoms. The methods of the present invention allow prediction and early treatment of patients before having advanced-stage disease,
[0054] In one embodiment, a patient is treated with a maximal cytoreduction strategy. In one embodiment an alternative treatment of a CLL indicated as having an adverse outcome is treated with this strategy. This strategy involves using a combination regimen to achieve a durable complete remission. On the basis of previously described randomized clinical trial, FCR is a popular choice for induction therapy (Haliek M, et al. : Lancet 376 (9747): 1164-74, 2010). The combination of BR (bendamustine + rituxirnab) is another regimen used for this strategy (Fischer K, et al.: J Clin Oncol 29 (26): 3559-66, 2011; and lannitto E, et al : Br J Haematol 153 (3): 351 - 7, 201 1). Other combinations include R-CHOP (rituxirnab + cyclophosphamide + doxorubicin + vincristine plus prednisone) and R-CVP (eliminating doxorubicin from R-CHOP).
[0055] In one embodiment, a patient is treated with an avoidance of alkylators and purine analogues strategy. The goal of this strategy is to minimize the possibility that subclones with deleterious mutations, which are subsequently resistant to further therapy, will emerge. In one embodiment, the presence of subclones having any of the driver mutations of the present invention are treated with this alternative strategy. This strategy also avoids prolonged cytopenias and the recurrent, long-lasting, sometimes fatal, infections seen after therapy with these agents. Initial therapy with a monoclonal antibody, such as rituxirnab (in high doses or more frequent scheduling) with or without maintenance rituxirnab, is consistent with this strategy. The subsequent use of a B-cell receptor inhibitor, such as ibrutinib or idelalisib at relapse, is consistent with this approach. (Furman RR, et al. : N Engl J Med 370 (1 1): 997-1007, 2014, and Byrd JC, et al. : N Engl J Med 369 (1): 32-42, 2013), Avoidance of alkylators or purine
analogues might be particularly advantageous for younger patients, who must reduce the risk of toxicities for many future years. This strategy is also used to prevent the harmful side effects of alkylator or purine analogue-based therapy in patients of advanced age and with significant comorbidities. In one embodiment, an alternative treatment of a CLL indicated as having an adverse outcome is treated with this strategy.
[0056] In one embodiment, a patient is treated with a risk-adapted therapy. Prior to the present invention prognostic factors, such as the mutational immunoglobulin variable region heavy chain (IgVH), 17p~, or HQ-status, were used to help inform the choice of therapy. Patients with a better prognosis might receive more gentle therapy with fewer short-term and long-term side effects. This approach might also be applicable for older patients with multiple comorbidities. Patients with a worse prognosis might receive combination regimens that are used for maximal cytoreduction. The methods of the present invention may be used to further determine a prognosis,
[0057] Standard options for CLL treatment are roughly ordered by level of toxic effects, starting with the least toxic options. In one embodiment, the present invention provides for treating a patient in need thereof based on identification of cancer drivers that indicate an adverse outcome. In one embodiment, treatment is with a more toxic option. The standard options include the following:
1. Observation. Outside of the context of a clinical trial, treatment for asymptomatic or minimally affected patients with chronic lymphocytic leukemia (CLL) is observation. Because the rate of progression may vary from patient to patient, with long periods of stability and sometimes spontaneous regressions, frequent and careful observation is required to monitor the clinical course.
2. Rituximab. Rituximab is a murine anti-CD20 monoclonal antibody (Mavromatis B, et ai. : J Clin Oncol 21 (9): 1874-81, 2003; O'Brien SM, et al. : J Clin Oncol 19 (8): 2165-70, 2001; Byrd JC, et al. : J Clin Oncol 19 (8): 2153-64, 2001.; Hainsworth ID, et al.: J Clin Oncol 21 (9): 1746-51 , 2003; and Castro JE, et al. : Leukemia 22 (11): 2048-53, 2008), When used alone, higher doses of rituximab or increased frequency or duration of therapy is required for comparable responses to those seen for other indolent lymphomas,
3. Ofatumomab. Ofatumomab is a human anti-CD20 monoclonal antibody (Wierda WG, et al. : J Clin Oncol 28 (10): 1749-55, 2010). A trial of 138 patients, who were previously
treated with fludarabine and alerntuzumab, showed overall response rates around 50% in patients refractory to fludarabine and with previous exposure to rituximab (Wierda WG, et al.: J Clin Oncol 28 (10): 1749-55, 2010; and Wierda WG, et ai.: Blood 118 (19): 5126-9, 2011).
Obinutuzumab. Obinutuzumab is a human anti-CD20 monoclonal antibody. In a randomized prospective trial (NCT01010061), 781 previously untreated patients with coexisting medical problems were randomly assigned to chlorambucil and obinutuzumab versus chlorambucil and rituximab versus chlorambucil alone (Goede V, et al. : N Engl J Med 370 (12): 1101-10, 2014). The median PFS was best for the obinutuzumab arm (26.7 months) versus the rituximab arm (16.3 months) versus chlorambucil alone (1 1.1 months ) (hazard ratio [HR], 0.18; 95% confidence interval [CI], 0, 13-0.24; P < .001) for obinutuzumab and chlorambucil versus chlorambucil alone; for rituximab and chlorambucil versus chlorambucil alone [HR, 0,44; 95% CI, 0.34-0.57; P < .001 ]. The 2- year OS was significantly improved for the obinutuzumab arm (91%) versus chlorambucil alone (80%) (HR, 0.41; 95% CI, 0.23-0.74, P = .002). Patients who received obinutuzumab did not have improved survival compared with those who received rituximab alone (Goede V, et al. : N Engl J Med 370 (12): 1101-10, 2014), Idelalisib. Idelalisib is an oral inhibitor of the delta isoform of the phosphatidylinositol 3- kinase, which is located in the B-cell receptor-signaling cascade. In a randomized, double-blind, prospective trial (NCT01539512), 220 patients treated mainly with fludarabine-based regimens and who had coexisting medical problems, such as renal dysfunction, received rituximab and idelalisib versus rituximab and placebo (Furman RR, et al: N Engl J Med 370 (11): 997-1007, 2014). With a median follow-up of less than 1 year, the PFS rate at 24 weeks favored the rituximab and idelalisib arm (93%) versus the rituximab and placebo arm (46%) (HR, 0.15; 95% CI, 0.08-0.28; P < .001), and the OS rate at I year was significantly better for the rituximab and idelalisib ami (92%) versus the rituximab and placebo arm (80%) (HR, 0.28, 95% CI, 0.09-0.86; P = ,02) (Furman RR, et al.: N Engl J Med 370 (1 1): 997-1007, 2014). A median PFS of 16 months was observed in a phase I trial of patients with heavily pretreated relapsed or refractory disease (Brown JR, et al.: Blood 123 (22): 3390-7, 2014).
6. Ibrutinib. Ibrutinib is a selective irreversible inhibitor of Bruton tyrosine kinase, a signaling molecule located upstream in the B-eell receptor-signaling cascade. Trials of previously untreated patients and of patients with relapsed or refractory CLL showed durable responses to the oral agent in phase I and II studies (Advani RH, et al. : J Clin Oncol 31 (1): 88-94, 2013; and O'Brien S, et al : Lancet Oncol 15 (1): 48-58, 2014). A phase fb -II trial (NCT01 105247) of 85 patients with relapsed or refractory CLL showed a 26-month PFS rate of 75% and included patients with 17p- or unmutated IgVH FISH testing. [ 12.Byrd JC, Furman RR, Coutre SE, et al.: Targeting BT with ibrutinib in relapsed chronic lymphocytic leukemia. N Engl J Med 369 (1): 32-42, 2013] Patients who discontinued ibrutinib early because of disease progression or drug intolerance had very poor outcomes, which were mainly attributable to very poor preexisting prognostic factors (Jain P, et al.: Blood 125 (13): 2062-7, 2015; and Maddocks KJ, et al.: JAMA Oncology 1 (1 ): 80-7, 2015).
[0058] A prospective randomized trial of 391 patients with relapsed or refractory CLL/SLL compared ibrutinib with ofatumumab. With a median follow-up of 9.4 months, the 12-month OS favored ibrutinib (90% to 81%, HR, 0.43; P = .005) (Byrd JC, et al. : N Engl J Med 371 (3): 213- 23, 2014). Similar outcomes were seen for patients whose disease was resistant to purine analogues or who had a chromosome 17p deletion.
7. Oral alkylating agents with or without corticosteroids. The French Cooperative Group on CLL randomly assigned 1 ,535 patients with previously untreated stage A disease to receive either chlorambucil or no immediate treatment and found no survival advantage for chlorambucil (Dighiero G, et al. : N Engl J Med 338 (21 ): 1506-14, 1998). A metaanalysis of six trials of immediate versus deferred therapy with chlorambucil (including the aforementioned trial by the French Cooperative Group) showed no difference in OS at 10 years (J Natl Cancer Inst 91 (10): 861-8, 3999).
8. Purine analogs. Several randomized trials have compared the purine analogs with chlorambucil; with cyclophosphamide, doxorubicin, and prednisone; or with cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) in previously untreated patients. All of these trials showed higher or equivalent response rates for the purine analog, and most showed an improvement in PFS; one reached significance in OS favoring f!udarabine (Rai KR, et al .: Blood 1 14 (22): A-536, 2009; Rai KR, et al : N Engl
J Med 343 (24): 1750-7, 2000; Robak T, Blood 96 (8): 2723-9, 2000; Johnson S, et al., Lancet 347 (9013): 1432-8, 1996; Leporrier M, et al. : Blood 98 (8): 2319-25, 2001 ; Eichhorst BF, et al. : Blood 1 14 (16): 3382-91, 2009; and Steurer M, et al. : Cochrane Database Syst Rev 3 : CD004270, 2006).
9. Bendamustine. Bendamustine is a cytotoxic agent with bifunctional properties of an alkylator and a purine analog (Leoni LM, et al. : Clin Cancer Res 14 (1): 309-17, 2008). In previously treated and untreated patients, bendamustine with rituximab has shown response rates around 70% to 90% (Fischer K, et al. : J Clin Oncol 29 (26): 3559-66, 2011; and lannitto E, et al: Br J Haematol 153 (3): 351-7, 201 1).
[0059] In a randomized comparison with chlorambucil in 319 previously treated patients, bendamustine showed a better response rate (68% vs. 31%, P < .0001) and PFS (21 .6 months vs. 8 months) with a median follow-up of 35 months (Knauf WU, et al. : J Clin Oncol 27 (26): 4378- 84, 2009). The German CLL Study Group compared bendamustine plus rituximab versus FCR as first-line therapy in patients with CLL who require therapy (Fischer K, et al. : J Clin Oncol 29 (26): 3559-66, 201 1).
10. Lenalidomide. Lenalidomide is an oral immunomodulatory agent with response rates over 50%, with or without rituximab, for patients with previously treated and untreated disease. Prolonged, lower-dose approaches and attention to prevention of tumor lysis syndrome are suggested with this agent (Chen CI, et al. : J Clin Oncol 29 (9): 1175-81, 201 1 ; Chanan-Khan A, et al.: J Clin Oncol 24 (34): 5343-9, 2006, Ferrajoli A, et al : Blood 111 (11): 5291-7, 2008; Strati P, et al. : Blood 122 (5): 734-7, 2013; Wendtner CM, et al : Leuk Lymphoma 53 (3): 417-23, 2012; Badoux XC, et al . : J Clin Oncol 31 (5): 584-91, 2013; and Moutouh-de Parseval LA, et al. : J Clin Oncol 25 (31): 5047, 2007).
1 1. Combination chemotherapy.
[0060] Other combination chemotherapy regimens include the following:
o Fludarabine plus cyclophosphamide plus rituximab (Lamanna N, et al. : J Clin Oncol 27 (4): 491-7, 2009, Foon KA, et al .: J Clin Oncol 27 (4): 498-503, 2009; Tarn CS, et al : Blood 1 12 (4): 975-80, 2008; and Badoux XC, et al.: Blood 117 (11): 3016-24, 2011),
o Fludarabine plus rituximab as seen in the CLB-9712 and CLB-901 1 trials (Woyach J A, et al.: J Clin Oncol 29 (10): 1349-55, 201 1).
o Fludarabine plus cyclophosphamide versus fludarabine plus cyclophosphamide plus rituximab (Robak T, et al . : J Clin Oncol 28 (10): 1756-65, 2010).
o Pentostatin plus cyclophosphamide plus rituximab as seen in the MAYO-MC0183 trial, for example (Kay NE, et al.: Blood 109 (2): 405-1 1, 2007, and Shanafelt
TD, et al.: Cancer 109 (1 1): 2291-8, 2007).
o Ofatumumab plus fludarabine plus cyclophosphamide (Wierda WG, et al. : Blood
117 (24): 6450-8, 2011).
o CVP: cyclophosphamide plus vincristine plus prednisone (Raphael B, et al. : J
Clin Oncol 9 (5): 770-6, 1991).
o CHOP: cyclophosphamide plus doxorubicin plus vincristine plus prednisone
(French Cooperative Group on Chrome Lymphocytic Leukemia. Leuk
Lymphoma 13 (5-6): 449-56, 1994).
o Fludarabine plus cyclophosphamide versus fludarabine as seen in the E2997 trial
[NCT00003764] and the LRF-CLL4 trial, for example (Flinn IW, et al.: J Clin
Oncol 25 (7): 793-8, 2007, Catovsky D, et al : Lancet 370 (9583): 230-9, 2007). o Fludarabine plus chlorambucil as seen in the CLB-90H trial, for example.
(Morrison VA, et al.: J Clin Oncol 20 (18): 3878-84, 2002).
Involved-field radiation therapy. Relatively low doses of radiation therapy will affect an excellent response for months or years. Sometimes radiation therapy to one nodal area or the spleen will result in abscopal effect (i.e., the shrinkage of lymph node tumors in untreated sites).
Alemtuzumab. Alemtuzumab, the monoclonal antibody directed at CD52, shows activity in the setting of chemotherapy-resistant disease or high-risk untreated patients with 17p deletion or p53 mutation (Moreton P, et al. : J Clin Oncol 23 (13): 2971 -9, 2005; Parikh SA, et al.: Blood 118 (8): 2062-8, 2011; Pettitt AR, et al. : J Clin Oncol 30 (14): 1647-55, 2012). In one embodiment, patients are treated if a cancer driver of the present invention is identified. As a single agent, the subcutaneous route of delivery for alemtuzumab is preferred to the intravenous route in patients because of the similar efficacy and decreased adverse effects, including less acute allergic reactions that were shown in some nonrandomized reports (Pettitt AR, et al. : J Clin Oncol 30 (14): 1647-55, 2012; Stilgenbauer S, et al. : J Clin Oncol 27 (24): 3994-4001 , 2009; Cortelezzi A, et al. :
Leukemia 23 (11): 2027-33, 2009; Osterborg A, et al.: Leukemia 23 (11): 1980-8, 2009, and Gritti G, et al.: Leuk Lymphoma 53 (3): 424-9, 2012).
[0061] In a randomized prospective study, 335 previously treated patients received intravenous alemtuzumab plus fiudarabine versus fiudarabine alone. With a median follow-up of 30 months, the combination of fiudarabine plus intravenous alemtuzumab had better PFS, with a median of 23.7 months versus 16.5 months (HR, 0.61; 95% CI, 0.47-0.80; P ------- .0003); and better
OS, with a median not reached, versus 52.9 months (HR, 0.65; 95% CI, 0.45-0.94; P = .021) (Elter T, et al. : Lancet Oncol 12 (13): 1204-13, 201 1).
14. Bone marrow and peripheral stem cell transplantations. Bone marrow and peripheral stem cell transplantations are alternative treatment options (Doney KC, et al.: Bone Marrow Transplant 29 (10): 817-23, 2002; Schetelig J, et al .: J Clin Oncol 21 (14): 2747- 53, 2003; Ritgen M, et al. : Blood 104 (8): 2600-2, 2004; Moreno C, et al.: J Clin Oncol 23 (15): 3433-8, 2005; houri IF, et al. : Cytotherapy 4 (3): 217-21 , 2002; Pavletic SZ, et al : J Clin Oncol 23 (24): 5788-94, 2005; and Dreger P, et al.: Blood 1 19 (21): 4851-9, 2012). In a prospective randomized trial, 241 previously untreated patients younger than 66 years with advanced-stage disease received induction therapy with a CHOP -based regimen followed by fiudarabine (Sutton L, et al.: Blood 117 (23): 6109-19, 2011). Complete responders (105 patients) were randomly assigned to undergo autologous stem cell transplantation (ASCT) or observation, while the other 136 patients were randomly assigned to receive dexamethasone, high-dose aracytin, and cisplatin reinduction followed by either ASCT or fiudarabine plus cyclophosphamide. Although the 3 -year EFS favored ASCT in complete responders, there was no difference in OS in any of the randomized comparisons.
15. Autologous T-cells directed at specific antigen targets. Autologous T-cells were modified by a ientiviral vector to incorporate antigen receptor specificity for the B-cell antigen CD19 and then infused into a previously treated patient (Porter DL, et al. : N Engl J Med 365 (8): 725-33, 2011). A dramatic response lasting 6 months has prompted larger trials of this concept. Ongoing clinical trials are testing the concept of T-cells directed at specific antigen targets with engineered chimeric-antigen receptors (termed CARs (Maus MV, et al. : Blood 123 (17): 2625-35, 2014).
[0062] In one embodiment, the present invention provides for treatment with a MEK inhibitor. A MEK inhibitor is a chemical or drug that inhibits the mitogen-activated protein kinase kinase enzymes MEK1 and/or MEK2. They can be used to affect the MAPK/ERK pathway which is often overactive in some cancers. The present invention provides for identification of CLL driver mutations indicating that treatment with MEK inhibitors should be performed. MEK inhibitors may be Trametinib (GSK1 120212), Selumetinib (Janne, Pasi A, et al. (2013). The Lancet Oncology 14 (1): 38-47) and is in phase III development in KRAS mutation positive NSCLC (SELECT-1,NCT01933932), Binimetinib (MEK 162), PD-325901, Cobimetinib or XLS 18, CI- 1040, or PD035901.
[0063] In another aspect, the present invention provides for novel targets for therapeutic intervention. Cancer drivers may be targeted with any agent that modulates the activity or expression of the target. The agent may be an antibody, a soluble polypeptide, a polypeptide agent, a peptide agent, a nucleic acid agent, a nucleic acid ligand, or a small molecule agent. The agent may target any of the genes or CNV's described herein. The nucleic acid agent may encode for RNAi, a Zinc finger nuclease (ZFN), a Transcription Activator-Like Effector Nuclease (TALEN), or a CRISPR/Cas system.
[0064] The present invention advantageously utilized large sequencing datasets of clinically informative samples to enable the discovery of novel cancer drivers and the network of relationships between the driver events and their impact on disease relapse and clinical outcome. Thus, the present advantageously allows improved clinical outcomes for patients suffering with
CLL,
[0065] Although the present invention and its advantages have been described in detail, it should be understood that various changes, substitutions and alterations can be made herein without departing from the spirit and scope of the invention as defined in the appended claims.
[0066] The present invention will be further illustrated in the following Examples which are given for illustration purposes only and are not intended to limit the invention in any way.
Examples
Example 1
[0067] Unbiased candidate CLL genes discovery. Applicants performed WES of CLL and matched germline samples, collected from 278 subjects enrolled on the CLL8 trial, with mean
read depth of 95.0 and 95.7, respectively {Supplementary Table 1). Consistent with previous CLL WES studies, Applicants detected a mean+/-SD rate of 21.5+/-7.9 silent and non-silent single nucleotide variants [sSNVs] and somatic insertions and deletions [sINDELs] per exome
1,3
[0068] Applicants inferred candidate cancer genes in CLL through implementation of MutSig2CV5'' . To maximize statistical sensitivity for driver detection5 Applicants combined the CLL8 cohort with two previously reported and non-overlapping WES cohorts1'', thereby increasing the size of the cohort to 538 CLLs. This cohort size is expected to saturate candidate CLL gene discovery for genes mutated in 5% of patients, and provides 94% and 61% power to detect genes mutated in 3% and 2% of patients, respectively5.
[0069] Applicants detected 44 putative CLL. driver genes, including 18 CLL mutated drivers that Applicants previously identified3, as well as 26 additional putative CLL genes {Fig. 1-2, Extended Data Fig. 1-2). In total, 33.5% of CLLs harbored mutation in at least one of these 26 additional genes. Targeted DNA sequencing as well as variant allele expression by RNAseq demonstrated high rates of orthogonal validation (Extended Data Fig. 3),
[0070] Of the newly identified putative cancer genes, some were previously suggested as CLL drivers in studies using other detection platforms. For example, the suppressor of MYC MGA (n=17, 3.2%), which Applicants detected as recurrently inactivated by insertions and nonsense mutations, was previously found to be inactivated through deletions8 and truncating mutations8,9 in high-risk CLL (Extended Data Fig. 4). A gene set enrichment analysis of matched RNAseq data revealed down-regulation of genes that are suppressed upon MYC activation in B-cellsl0. In addition to MGA, Applicants report two additional candidate driver genes that likely modulate MYC activity (PTPNll11 [n=7, 1.3%] and FUBP112 [n=9, 1.7%]), highlighting MYC-related proteins as drivers of CLL.
[0071] Another cellular process affected by novel CLL drivers is the MAPK-ERK pathway, with 8.7% of patients harboring at least one mutation in CLL genes in this pathway. These included mutations in RAS genes (NBAS, n 9 and KRAS n 14, totaling 4. 1 %); BRAF in 2 1 , 3.7%); or the novel putative driver MAP2K1 (n=12, 2%). This finding suggests therapeutic options including MAPK-ERK. pathway inhi bitors in CLL. Intriguingly, BRAF mutations in CLL did not involve the canonical hotspot (V600E) seen in other malignancies5,1 '14, but rather clustered heavily around the activation segment of the kinase domain (Fig. 2). This may hint at a
different mechanism of activity15'1", and has clinical implications, as BRAF inhibitors are thought to be less effective for non-canonical BRAF mutations' 7,18 and may be more effectively targeted by MEK inhibitors (Pratilas, C. A. et al. Genetic predictors of MEK dependence in non- small cell lung cancer. Cancer Res 68, 9375-9383, doi : 10.1 158/0008-5472.CAN-08-2223 (2008))..
[0072] In addition to highlighting novel cellular processes and pathways affected in CLL, many of the 26 additional CLL genes more densely annotated pathways or functional categories previously identified in CLL19, including R A processing and export (FUBPI, XP04, ESWR1, NXFI), DNA damage (CHEK2, BRCC3, ELF420 and DYRK1A21), chromatin modification (ASXLI, HIST1H IB, BAZ2B, FKZF3) and B cell activity related pathways (TRAF2, TRAF3, CARD11).
[0073] Applicants discovered a number of putative CLL drivers previously unrecognized in human cancer. In a first example. Applicants found that RPS15 was recurrently mutated (n==:23, 4.3%), with mutations localized to the C -terminal region (Fig. 2) at highly conserved sites (median conservation score of 94/100). This component of the S40 ribosomal sub unit, has not been extensively studied in cancer, although rare mutations have been identified in Diamond- Blackfan anemia22. A gene set enrichment analysis revealed upregulation of gene sets related to adverse outcome in CLL as well as immune response gene sets. In another example of a previously unrecognized cancer gene, Applicants identified recurrent L162R substitutions (n=l l , 2.0%) in IKZF3, targeting a highly conserved amino acid (93/100 conservation score). This gene is a key transcription factor in B cell development'3, and its upregulation has been associated with adverse outcome24,25.
[0074] In addition to sSNVs and sINDELs, Applicants characterized somatic copy number variations (sCNVs) directly from the WES data (Extended Data Fig. 5, Supplementary Table 5). When Applicants accounted for all 55 identified driver events - including non-silent sSNVs and sINDELs in putative CLL genes (n=44), and recurrent sCNVs (n=l 1)— 91.1% of CLLs contained at least one driver. Moreover, 65.4% of CLLs now harbor at least 2 drivers, and 44.4% at least 3 drivers, compared with 55.9% and 3 1.8% were Applicants to exclude the 26 additional CLL genes.
Supplementary Table 1: Patients characteristics ©f the 278 patients that rovided s m les m part of GCLLSG-CLL8 trial.
Respoader versus non-response/missmg
S«p|>!e#*etttary Tafeie 5: (.'¾>?«ρ, trfcon between $os« c copy
deiee k i with WES vs. ISH * oge.»efks
Example 2
[0075] Drivers and CLL characteristics. The larger cohort size also provided statistical power to examine associations between genetic alterations and key CLL features. First, Applicants examined whether mutations differed between IGHV mutated and unmutated
subtypes, the two main subtypes of CLL. In agreement with the relative clinical aggressiveness of IGHV unmutated CLL, most drivers were found in a higher proportion in this subtype (Extended Data Fig. 6 A). Only three driver genes were enriched in the IGHV mutated CLL (iii?/(13q), MYD88, ( 7/7)2), suggesting a role for these specific alterations within the oncogenic process of this subtype.
[0076] Second, since therapy could lead to selection of particular driver events, Applicants examined the 33 samples (6.2%, none enrolled on CLL8) that had received therapy prior to sampling. Prior treatment was associated with enrichment in TP53 and BIRC3 mutations, del(np) and de!(l lq) as previously indicated26, as well as in mutated DDX3X and MAP2K1, suggesting their selection by therapeutic interventions (Extended. Dat Fig. 6B).
[0077] Third, Applicants examined whether coherent patterns of co-occurrence of driver events were evident, limiting our analysis to the 31 drivers with >10 affected patients. Of 465 possible pairs, 1 1 combinations had statistically significant high or low co-occurrence (Extended Data Fig. 6C-D). As expected, a high degree of co-occurrence was found between mutated TP53 and (. ('/( 1 7p). and between mutated AIM. and dei(l lq). Both mutated AIM and de!(l lq) significantly co-occurred with amp(2p), and associations between presence of tri(\2) with mutated BIRC3 and with mutated BCOR were also found. A significantly low rate of cooccurrence was seen between tfe/(13q) and /·/'{ 12).
[0078] Fourth, Applicants examined the temporal sequence of driver acquisition in the evolutionary history of CLL, To do this, Applicants computed the cancer-cell fraction (CCF) of each mutation across the 538 samples, and identified mutations as either clonal or subcional2 ' (58.1% of mutations classified as subcional). Both clonal and subcional sSNVs were similarly dominated by C>T transitions at C*pG sites {Extended Data Fig. 7).
[0079] Applicants first classified driver events likely acquired earlier or later in the disease course based on the proportion of cases in which the driver was found as clonal {Fig. 3Λ). This large dataset further enabled the inference of temporal relationships between pairs of drivers. Applicants systematically identified instances in which a clonal driver was found together with a subcional driver within the same sample, as these pairs reflect the acquisition of one lesion (clonal) followed by another (subcional), providing a temporal 'edge' leading from the former to the latter2,8'29. For each driver, Applicants calculated the relative enrichment of out-going edges compared to in-going edges to define early, late and intermediary drivers (Supplementary Table
7). For 23 pairs connected by at least 5 edges. Applicants further established the temporal relationship between the two drivers in each pair, and thereby constructed a temporal map of the evolutionary trajectories of CLL {Supplementary Table 8, Fig. 3B)" . This network highlights sCNVs as the earliest events with two distinct points of departure involving del(\3q) and //· ( 12). It further demonstrates an early convergence towards de!(l lq) and substantial diversity in late drivers. Finally, this analysis suggests that in the case of the tumor suppressor genes ^ Z and BIRC3, copy loss precedes sSNVs and sINDELs in bialleiic inactivation.
Su plementar Table 7; Temporal order of somatic mutation acquisitions -■ clas»ifyi»g drivers as ea ly vs. late.
These table includes all driver events (recurrent sCNVs and candidate CLL gene non-silent mutations), classifying the driver events based on the relative enrichment of out-degrees vs. in- degrees as early (O<0.2 and number of out-degrees > in-degrees). late {Q<Q,2 and number of out-degrees < in-degrees) and intermediate or not powered (£>>0.2. Inter./not powered). Out- degrees are defined as instances in which the driver event is clonal and found in the same CLL with another driver event that is subclonal. in-degrees are defined as instances in which the driver event is subclonal and found in the same CLL with another driver event that is clonal.
Table 7 a: Results in n = 501 treatment naive patients (4 patients with unknown status of prior therapy were excluded from the analysis as well):
Driver O-value occurin-degrees out-degrees classification
event rences
del 13q L22E-23 233 35 1 83 Early
tri12 9.44E-22 67 i 81 Early
ATM 2.94E-13 76 89 14 Late
BI. C3 2.05E-07 15 27 0 Late
dell I q 3.93E-04 103 42 90 Early
del20p 2.24E-03 6 0 1 Early
FBXW7 1.53E-02 10 10 0 Late
MAP2K1 2.39E-02 8 9 0 Late
NRAS 2.39E-02 8 9 0 Late
KRAS 3.49E-02 14 1 1 1 Late
BAZ2A 3.58E-02 9 8 0 Late
MYD88 3.58E-02 14 0 8 Early
CARD! 1 4.64E-02 7 10 1 Late
Mi A 4.64E-02 15 16 4 Late
ΖΜΎΜ3 4.74E-02 10 12 Late
TP53 6.90E-02 29 26 1 1 Late
amp2p 9.54E-02 45 44 25 Late
del6q2.1 L60E-01 16 1 6 Late
DYR 1 A 1 .93E-01 7 7 I. Late
FAM50A 1.93E-01 5 7 1 Late
CHEK2 3.12E-01 5 1 6 Inter . not powered
TRAF3 3.12E-01 4 4 0 Inter./not powered
BRAF 3.35E-01 19 12 5 inter./not powered
1RF4 3.35E-01 10 9 3 Inter./not powered
NOTCH 1 3.38E-01 37 25 15 Inter . not powered
ampSq 3.531.-01 12 10 4 1 nter . not powered
MED 12 3.53E- I "7 "7 2 I liter .n ot powered
TRAF2 3.53E-01 6 7 · Inier./not powered
ΡΪΡΝΠ 4.15E-01 6 5 1 Inter .not powered
ΡΪΜΙ 4.44E-01 3 0 i ter, /not powered tril 4.44E-01 6 0 3 Inter./not powered
ASXLi 4.82E-01 5 6 2 1 nter. /not powered
EL.F4 4.82E- 1 7 6 Inter./not powered
RPS15 5.29E-01 21 16 1 Inier./not powered
SF3B1 5.32E-01 103 72 60 Inter./not powered
NXFI 5.G7E-01 7 4 1 inier./not powered
POTl 5.67E-01 33 20 27 Inter./not powered
GNB1 6.56E-0I 5 5 2 Inter./not powered
HISTIH!E 7.16E-01 7 3 6 Inter./not powered
BRCC3 7.55E-01 5 7 4 1 nter ./n ot powered
IKZF3 7.79E-01 ii 5 S Inter./not powered dell 8p 8.24Ε-0Ϊ 12 10 7 Inter./not powered
EGR2 8.67Ε-ΘΙ 16 10 13 Inter./not powered dell7p 9.10E-01 24 15 18 Inter./not powered
FUB 1 9. EE-0I 9 8 6 inter./not powered
CHD2 9.61E-01 23 9 7 I nter in ot poweed
XPOI 9.95E-01 22 13 15 inter./not powered
BCOR L OE+00 10 7 6 Inter./not powered
IGLL5 LOOE+00 11 6 7 Inter./not powered
DDX3X 1. OE+00 10 4 4 Inter./not powered delSp LOOE+00 16 10 10 Inter, /n ot pow ered
EWSRJ 1.OOE+00 4 1 I nter ./not powered
MIST 1 Ml B LOOE+00 4 2 Inier./not powered
SA.M.HD 1 1.OOE+00 10 6 6 Inter./not powered
X.P04 LOOE+00 7 5 5 Inter./not powered
Table 7b: Results in n = 229 treatment naive patients with IGHV unmntated CLL:
EGR.2 8.54E-01 14 9 12 inter./not powered
XPO i 8.54E-01 20 10 13 inter./not powered
RPS I 5 8.54E-01 1.9 14 1 1 Inter ./not powered
BRCC3 8.79E-01 4 5 3 inter./not powered
J.KZF3 8.9 ! E-01 8 4 6 Inter./not powered
DDX3.X l .OOE+OO 8 3 4 Inter./not powered
SAMHD S L00E÷00 7 3 4 Inter./not powered
SF3B I 1..00F 00 60 41 42 Inter./not powered
BCOR 1 .OOE+00 8 7 6 Inter./not powered amp8q LOOE+00 5 1 2 Inter./not powered
FAM50A :i .ooE+oo ! 0 1 Inter./not powered
FUBPl i .OOE+00 6 5 5 inter./not powered
TRAF2 1 .OOE+00 4 3 - Inter./not powered
Table 7c: Results in n ~ 245 treatment naive patients with IGHV mutated CLL:
Supplementary Table 8: Temporal order of so a tie oMsteikso acquisitions ~ pair fee data.
These tables includes all pairs of driver events (dl, d2) that had at least 5 cases in which the two drivers were detected in the same CLL sample, but one of the drivers is clonal and the other is subclonal. A two-tailed binomial test is performed to test whether the pairings are found to be in one order more frequently than the other (i.e., dl c!2 > d2 dl ). A multi-hypothesis correction is then applied and the table lists all hypotheses tested.
Table 8a: Pairing in a = 501 treatment naive patients ( 4 patients with unknown status of prior therapy were excluded f om the analysis as well):
Ordering P- value £?-value No. orderings No. opposite
(clonal - orderings
subclonal) (subclonal
clonal)
dell3q->ATM 1.54E-08 3.53E-07 31 1
dell1q->ATM 2.10E-05 2.41E-04 20 1
del Bq->auip2p 6.10E-05 4.68E-04 15 0
dell3q->SP3Bl 3.24E-04 1.86E-03 27 6
dell lq~>amp2p 9.77E-04 4.49E-G3 14 1
tril2~>BlRC3 3.91E-03 E50E-02 9 0
tril2->delllq 3.12E-02 1.03E-01 6 0
de!llq->BlRC3 6.25E-02 1.20 !.-()! 5 0
dell3q->del6cj21 6.25E-02 1.20E-01 5 0
dell3q->MGA 6.25E-02 1.20E-01 5 0
trii2->FBXW7 6.25E-02 1.20E-01 5 0
tril2-> RAS 6.25E-02 1.20E-01 5 0
amp2p- ATM 7.03E-02 1.24E-01 1
dell q->de!11q 9.31E-02 1.53E-01 16
dell3q->POTl 1.25E-01 L80E-01 6 1
deil3q->TP53 1.25E-01 1.SOE-01 6 1
dell!q->SF3Bl 2.67E-01 3.61E-01 9 4
de113q-X_el17p 3.75E-01 4.54E-01 4 1
delI3q->NOTCHl 3.75E-01 4.54E-01 4 1
SF3BI->ATM 5.08E-01 5.S4E-01 6 3
deU7p->TP53 6.88E-01 7.53E-01 4
XP01->ddl3q 1.00E+(M) LOOE+OO 4 3
POTl->deiiiq LOOE+OO 1.00E+00 3
Table 8b: Pairing in n - 229 treatment naive patients with /GHFunmutated CLL, Patients with unknown IGHV status were excluded from the analysis:
Table 8c: Pairings in n = 245 treatment naive patients with IGHV mutated CLL. Patients with unknown IGHV " status were excluded from the analysis:
Example 3
[0080] Impact of presence and clonality of CLL drivers on clinical outcome. Applicants examined whether presence of any of the drivers detected in at least 10 of the 278 pre-treatment CLL8 samples was associated with impact on clinical outcome (Fig. 4A, Extended Data Fig. 8- 9; the genomics analysis team was blinded to the clinical outcome data). Previous investigations suggested an impact for 7 CLL genes (SF3HI. A TM, TPS '3, XPOl, EGR2, POTl and BIRC3)30'33.
Applicants found shorter progression-free survival (PFS) associated only with TP53 and SF3B1 mutations. Of the newly identified recurrent lesions evaluated (MGA, BRAF and RPS15), Applicants observed a shorter PFS with mutated RPS15 (Bonferroni P = 0.024).
[0081] Presence of a detectable pre-treatment subclonal driver has been previously associated with shorter remissions in patients treated with heterogeneous therapies3. In the CLLS cohort, Applicants again found that the presence of a pre-treatment subclonal driver was associated with a significantly shorter PFS (hazard ratio (HR) 1.6 [95%CI 1.2-2.2, P = 0.004). This association remained significant in both the FC and FCR treatment arms (Fig. ¥11), with a non-significant trend when IGHV mutation status was added to a muitivariable model in addition to the treatment arm (1.3 [0,9-1 ,9], P=0.102). Applicants also note a trend towards shorter OS in CLLs with a subclonal driver (P = 0.151 ).
Example 4
[0082] Clonal evolution at disease relapse.
[0083] To define clonal evolution in disease relapse, Applicants performed WES on matched samples collected at the time of relapse from 59 of 278 CLL8 subjects (Supplementary Table 9), Applicants observed large clonal shifts between pre-treatment and relapse samples in the majority of cases (57 of 59), thus demonstrating that CLL evolution after therapy is the rule rather than the exception (Fig. 5A). The relapse clone was already detectable in pre-treatment WES in 18 of 59 (30%) cases, demonstrating that the study of pre-treatment diversity anticipates the future evolutionary trajectories of the relapsed disease34. By targeted deep sequencing, Applicants detected relapse drivers in 1. 1 of the 41 of pre-treatment samples in which WES did not detect the relapse driver. In 7 of these 1 1 CLLs, at least one relapse driver was detected in the pretreatment sample.
[0084] Applicants further compared the pre-treatment and relapse CCF for each driver, and observed three general patterns. First, //· ( ! 2), d.el(\3q) and afe/fl l q), suggested as early drivers (Fig. SB), tended to remain stably clonal despite marked, often branched, evolution (Fig. SB [CLL cases: GCLL-1 15, 307], Fig. 5C-top; Extended Data Fig. 10). This confirms that these are indeed early events likely shared by the entire malignant population . Second, TP53 mutations and dei(l7p) demonstrated increases in CCF upon relapse, suggesting a fitness advantage under therapeutic selection (Fig. SB [GCLL-27, Fig. SC-middle\). The novel driver 1KZF3 increased in
CCF in 3 of 4 relapse cases (and remained clonal in the fourth), supporting that these mutations likely enhance fitness. Third, mutations in SF3B1 and ATM, identified as a temporally intermediate or late drivers, seemed just as likely to fall in CCF as they were to rise (Fig. 5C- Bottom). These results suggest that within this therapeutic context such mutations do not provide as strong of a fitness advantage as TP53 disruption. In addition, Applicants observed 9 instances each of multiple distinct alleles of Λ ΊΜ and SF3BI mutations within the same CLL, (e.g., GCLL-307 in Fig. SB), indicating convergent evolution of these late-occurring CLL drivers.
[0085] This series also informs us regarding the mutagenesis of the tumor suppressor genes TP53 and ATM, where biallelic inactivation is common. In the case of ATM, Applicants typically find a fixed clonal dei(l lq223) and subclones harboring sSNVs affecting the other allele that shift in CCF over time (e.g., GCLL -307). Applicants confirmed that the breakpoints of sCNVs in matched relapse and pre-treatment samples were highly consistent, likely representing the same deletion event. These data thus suggest that mono-allelic ATM deletion provides a fitness advantage that enables the expansion of the malignant population with subsequent growth of multiple co-existing clones that harbor a second hit in the remaining allele. Thus while a biallelic lesion is clearly selected for (Extended Dat Fig. 6C), the longitudinal data support the temporal analysis (Fig. 3B) in which del(l lq) precedes AIM. mutations, reflecting the higher likelihood of a focal copy number loss compared with a deleterious point mutation''3,36. In contrast, Applicants consistently observed a concordant rise of <afe/(17p) and ΊΡ53 mutations in all 12 CLLs harboring both of these events, and none of these cases exhibited multiple shifting TP53 sSNVs/sINDELs. These observations suggest that a true biallelic inactivation of ΊΡ53 is required, and indeed, across the 538 CLL samples, the odds ratio for co-occurrence of i¾?/(17p) and TP53 mutation was far greater than the odds ratio for co-occurrence of del(\ lq) and ATM mutation (97.22 vs. 10.99, respectively). These observations are in agreement with a recent analysis that suggested that with the exception of a few genes such as TP53, tumor suppressor genes in sporadic cancers are haploinsufficient to begin with, and that the second hit only further builds on this fitness advantage'7.
Supplementa Table 9: Beseriptio.® o.f patients for whkh matched pre-breateeat ami rela se sampler were an lyze ..
Example 5
[0086] CONCLUSIONS. This study of WES in CLL enabled a comprehensive identification of putative cancer genes in CLL, generating novel hypotheses regarding the biology of this disease, and identifying previously unrecognized putative CLL drivers such as RPS15 and IKZF3, The detailed characterization of the compendium of driver lesions in cancer is of particular importance as Applicants strive to develop personalized medicine, as driver genes
may inform prognosis (e.g., RPS15 mutations) and identify lesions that may be targeted by therapeutic intervention (e.g., MAPK pathway mutations and specifically the unexpected enrichment for non-canonical BRAF mutations). Through the inclusion of samples collected within a landmark clinical trial with mature outcome data. Applicants could further study of the impact of genetic alterations in the context of the current standard-of-care front line therapy. As targeted therapy is rapidly transforming the treatment algorithms for CLL, future studies will be required to reexamine these associations in this context38.
[0087] An important benefit of the larger cohort size is the enhanced ability to explore relationships between driver lesions based on patterns of their co-occurrence. Focusing on temporal patterns of driver acquisition - based on the distinction between clonal versus subclonal alterations in a cross-sectional analysis - Applicants derived a temporal map for the evolutionary history of CLL. In the context of relapse after first-line fludarabine based therapy, Applicants note highly frequent clonal evolution, and that the future evolutionary trajectories were already anticipated in the pre-treatment sample in one third of cases with WES.
[0088] This study provides a glimpse of some of the anticipated fruits of the application of novel genomic technologies to growing cohort sizes across ieukemias: the continued discovery of novel candidate cancer genes, the deeper integration of genetic analysis with standardized clinical information (collected within clinical trials) to inform prognosis and therapy, and the ability to delineate the complex network of relationships between cancer drivers in the history and progression of the malignant process.
References
1 Quesada, V. et al. Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia. Nat Genet 44, 47-52, doi: 10.1038/ng. l032 (2012).
2 Puente, X. S. et al Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature 475, 101 - 105, doi: 10.1038/naturel 0113 (2011).
3 Landau, D. A. et al. Evolution and impact of subclonal mutations in chronic lymphocytic leukemia. Cell 152, 714-726, doi : ] 0.1016 j . cell .2013.01.019 (2013).
Schuh, A. el al. Monitoring chronic lymphocytic leukemia progression by whole genome sequencing reveals heterogeneous clonal evolution patterns. Blood 120, 4191-4196, doi: 10.1 182/blood-2012-05-433540 (2012). Lawrence, M. S. et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 505, 495-501, doi: 10.1038/naturel2912 (2014). Hallek, M. et al. Addition of ntuximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: a randomised, open-label, phase 3 trial. Lancet 376, 1164- 1174, doi : 10.1016/SO 140-6736( 10)61381-5 (2010). Lawrence, M. S. et al. Mutational heterogeneity in cancer and the search for new cancer- associated genes. Nature 499, 214-218, doi: 10.1038/naturel2213 (2013). Edelmann, J. et al. High-resolution genomic profiling of chronic lymphocytic leukemia reveals new recurrent genomic alterations. Blood 120, 4783-4794, doi: 10.1182/blood- 2012-04-423517 (2012), De Paoli, L. et al. MGA, a suppressor of MYC, is recurrently inactivated in high risk chronic lymphocytic leukemia. Leuk Lymphoma 54, 1087-1090, doi: 10.3109/10428194.2012.723706 (2013), Schlosser, I. et al. Dissection of transcriptional programmes in response to serum and c- Myc in a human B-eeli line. Oncogene 24, 520-524, doi : 10. 1038/sj one. 1 208198 (2005). Jiang, X, et al. Critical role of SHP2 (PTPN11) signaling in germinal center-derived lymphoma, Haematologica 99, 1834-1845, doi : 10.3324/haematol.2014.106401 (2014). Zhang, J. & Chen, Q. M. Far upstream element binding protein 1 : a commander of transcription, translation and beyond. Oncogene 32, 2907-2916, doi : 10.1038/onc.2012.350 (2013). Tiacci, E. et al. BRAF mutations in hairy-cell leukemia. N Engl J Med 364, 2305-2315, doi : 10, 1056/ EJMoal 014209 (2011). Brastianos, P. K. et al. Exome sequencing identifies BRAF mutations in papillary craniopharyngiomas. Nat Genet 46, 161-165, doi: 10.1038/ng.2868 (2014).
Heidorn, S, J. et al. Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell 140, 209-221 , doi: 10.1016/j . cell.2009.12.040 (2010). Cancer Genome Atlas Research, N. Integrated genomic characterization of papillary thyroid carcinoma. Cell 159, 676-690, dor 10 l O Kvj .cci! 201 09 050 (2014), Yang, H. et al RG7204 (PLX4032), a selective BRAFV600E inhibitor, displays potent antitumor activity in preclinical melanoma models. Cancer Res 70, 5518-5527, doi : 10.1158/0008-5472.C AN-10-0646 (2010), Jebaraj, B. M. et al. BRAF mutations in chronic lymphocytic leukemia. Leuk Lymphoma 54, 1177-1182, doi: 10.3109/10428194,2012,742525 (2013). Landau, D. A. & Wu, C. J. Chronic lymphocytic leukemia: molecular heterogeneity revealed by high-throughput genomics. Genome Med 5, 47, doi : 10.1 186/gm451 (2013). Sashida, G. et al. ELF4/MEF activates MDM2 expression and blocks oncogene-induced pl6 activation to promote transformation. Mol Cell Biol 29, 3687-3699, doi : 10.1128/MCB .01551-08 (2009). Park, J. et al. DyrklA phosphorylates p53 and inhibits proliferation of embryonic neuronal cells. J Biol Chem 285, 31895-31906, doi: 10.1074/jbc.M110.147520 (2010). Gazda, H. T. et al Ribosomal protein L5 and Ll l mutations are associated with cleft palate and abnormal thumbs in Diamond-Blackfan anemia patients. Am J Hum Genet 83, 769-780, doi: 10.1016/j .ajhg.2008.1 1.004 (2008), Ferreiros-Vidal, I. et al. Genome-wide identification of Ikaros targets elucidates its contribution to mouse B-ceil lineage specification and pre-B-cell differentiation. Blood 121, 1769-1782, doi : 10.1182/blood-2012-08-450114 (2013). Billot, K. et al Deregulation of Aiolos expression in chronic lymphocytic leukemia is associated with epigenetic modifications. Blood 117, 1917-1927, doi: 10.1182 blood- 2010-09-307140 (201 1), Nuckel, H. et al. The I ZF3 (Aiolos) transcription factor is highly upreguiated and inversely correlated with clinical progression in chronic lymphocytic leukaemia. Br J Haematol 144, 268-270, doi : 10.1111/j . l 365-2141.2008.07442.x (2009).
Bea, S. et al Genetic imbalances in progressed B-cell chronic lymphocytic leukemia and transformed large-ceil lymphoma (Richter's syndrome). Am J Pathol 161, 957-968, doi: 10.1016/80002-9440(10)64256-3 (2002). Carter, S. L. et al. Absolute quantification of somatic DNA alterations in human cancer. Nature Biotechnol 30, 413-421, doi: 10.1038/nbt.2203 (2012). Papaemmanuil, E. et al. Clinical and biological implications of driver mutations in myelodysplasia syndromes. Blood 122, 3616-3627; quiz 3699, doi : 10.1182/blood-2013- 08-518886 (2013). Wang, J. et al. Tumor evolutionary directed graphs and the history of chronic lymphocytic leukemia. Elife 3, doi : 10.7554/eLife.02869 (2014). Stilgenbauer, S. et al. Gene mutations and treatment outcome in chronic lymphocytic leukemia: results from the CLL8 trial. Blood 123, 3247-3254, doi: 10.1182/blood-2014- 01-546150 (2014). Rossi, D. et al. Integrated mutational and cytogenetic analysis identifies new prognostic subgroups in chronic lymphocytic leukemia. Blood 121, 1403-1412, doi: 10.1 182/blood- 2012-09-458265 (2013). Damm, F. et al. Acquired initiating mutations in early hematopoietic cells of CLL patients. Cancer Discov 4, 1088-1 101, doi: 10.1158/2159-8290.CD-14-0104 (2014). Winkelmann, N. et al. Low frequency mutations independently predict poor treatment- free survival in early stage chronic lymphocytic leukemia and monoclonal B-cell lymphocytosis, Haematologica 100, e237~239, doi: 10.3324/haematol.2014.120238 (2015). Puente, X. S. & Lopez-Otin, C. The evolutionary biography of chronic lymphocytic leukemia, Nature Genet, doi: 10.1038/ng.2556 (2013). Nowak, M. A. et al. The role of chromosomal instability in tumor initiation. PNAS 99, 16226-16231, doi: 10.1073/pnas.202617399 (2002). Bozic, I. et al. Accumulation of driver and passenger mutations during tumor progression. PNAS 107, 18545-18550, doi : 10.1073/pnas.1010978107 (2010).
37 Davoli, T. et al Cumulative haploinsufficiency and triplosensitivity drive aneuploidy patterns and shape the cancer genome. Cell 155, 948-962, doi : 10.1016/] . cell.2013.10.011 (2013).
38 Hallek, M. Chronic lymphocytic leukemia: 2015 Update on diagnosis, risk stratification, and treatment. Am JHematol 90, 446-460, doi: 10.1002/ajh.23979 (2015).
39 Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415-423 , doi : 0, 1038/nature 12477 (2013).
40 Vogel stein, B. et al. Cancer genome landscapes. Science 339, 1546-1558, doi: 10.1 126/science.1235122 (2013).
Methods
[0089] Human samples: Heparinized blood was obtained from patients enrolled on the prospective, randomized, open-label CLL8 trial1 before the first cycle of treatment. Sample selection was based on availability, and the baseline characteristics of the cohort of patients studied with WES are largely reflective of the baseline characteristics of the entire CLL8 cohort {Supplementary Table 1). All patients had a diagnosed CLL according to WHO criteria confirmed by flow cytometry and were in Binet Stage C or Binet A or B with need for treatment as defined by the study inclusion criteria1. Peripheral blood mononuclear cells (PBMC) from patients were isolated by Ficoll density gradient centrifugation. Immuno-magnetic tumor cell enrichment via CD 19 was performed on all baseline pretreatment samples (Midi MACS, Miltenyi Biotec, Bergisch Gladbach, Germany) achieving a separation of PBMCs into a CD19- positive tumor sample and a CD19-negative normal sample with a purity of >95% by flow cytometry. For some samples, the source of matched normal tissue was PBMC collected following chemoimmunotherapy when the samples were evaluated by flow cytometry as minimal residual disease-negative (Kiel Laboratory, Germany). Samples were used fresh or cryopreserved (with FBS with 10% DMSO) and stored in vapor-phase liquid nitrogen until the time of analysis.
[0090] Established CLL prognostic factor analysis: Analyses of genomic aberrations and immunoglobulin heavy-chain variable (IGHV) homology were performed in the central reference
laboratory of the German CLL Study Group (GCLLSG) in Ulm, Germany. Unmutated IGHV was defined as greater than or equal to 98% homology to the closest germline match analyzed via DNA sequencing. Greater than 20% ZAP-70 expression was considered positive (high-risk)2. Cytogenetics were evaluated by FISH for the most common CLL abnormalities (tfe/(1.3q), tri(\2), del{\ \q), (Ιβί(Πρ), rearrangements of chromosome 14) (probes from Vysis, Des Plaines, IL). Samples were scored positive for a chromosomal aberration based on consensus cytogenetic scoring3. Statistical analysis considered the hierarchical Doehner classification \
[0091] DNA quality control: Applicants used standard Broad Institute protocols as previously described5,6. Tumor and normal DNA concentration were measured using PicoGreen® dsDNA Quantitation Reagent (Invitrogen, Carlsbad, CA). A minimum DNA concentration of 5 ng/μΐ was required for sequencing. All Illumina sequencing libraries were created with the native DNA. The identities of ail tumor and normal DNA samples were confirmed by mass spectrometric fingerprint genotyping of 95 common SNPs by Fluidigm Genotyping (Fluidigm, San Francisco, CA).
[0092] Whole exome sequencing (WES): Informed consent for genomic analyses of patients' samples was obtained prior to the initiation of sequencing studies. The study was approved by the Ulm University Ethics Committee, IRC/EC number 138/03. DNA was extracted and purified from CD 19+ and CD 19- fractions of PBMC for tumor and matched germline DNA, respectively, using the Qiagen all-prep kit (Quiagen, Hilden, Germany) according to its unmodified protocol. Libraries for whole exome sequencing were constructed and sequenced on either an Illumina HiSeq 2000 or Illumina HiSeq 2500 using 76 bp paired-end reads. Details of whole-exome library construction have been described elsewhere '. Standard quality control metrics, including error rates, percentage-passing filter reads, and total Gb produced, were used to characterize process performance before downstream analysis. Note that due to a change in the Agilent capture bait set, the NOTCH 1 hotspot was not covered in samples GCLL-199 through GCLL-313, which included 10 samples with NOTCH! c.7544_7545delCT deletions by Sanger sequencing8. CLL samples found to harbor NOTCH! mutations by Sanger sequencing were subsequently submitted to targeted sequencing with Illumina TruSeq Custom Amplicon library, and sequenced on an Illumina MiSeq with a mean coverage depth of 1332X. In the relapse samples, samples with NOTCH 1 mutations by Sanger sequencing (GCLL-0146-T-02, GCLL-0192-T-02, GCLL-0049-T-02, GCLL-0208-T-02) were also submitted to targeted
sequencing. In addition to samples from patients included in the CLL8 clinical trial, Applicants have analyzed 157 WES samples from the cohort Applicants have previously published5'. The sequencing reads were realigned to hgl9 and all downstream analysis was done using the same methods as with the sequencing data for the CLL8 cohort. Finally, previously published WES data for 103 matched CLL and germline DNA samples were downloaded with permission from the European Genome-Phenome Archive10, The raw sequencing reads were processed in identical fashion to the in house produced WES libraries. New WES data is deposited in dbGaP (phs000922.vl .pl).
[0093] Identification of somatic mutations: Output from Iflumina software was processed by the "Pi card" data processing pipeline to yield BAM files containing aligned reads (bwa version 0,5.9, to the NCBI Human Reference Genome Build hgl 9) with well-calibrated quality scores 5, 11. From the sequencing data, somatic alterations were identified using a set of tools within the "Firehose" pipeline, developed at the Broad Institute (www.broadinstitute.org/cancer/cga). The details of the sequencing data processing have been described elsewhere5,6. Somatic single nucleotide variations (sSNVs) were detected using MuTect (Firehose version vl3112); somatic small insertions and deletions were detected using an improved version (manuscript in preparation, Cibulskis et al.,) of IndelocatorJ. The primary improvement is implementation of local reassembly, which results in more accurate allele fraction estimation. Following our standard procedure, Applicants filter sSNVs and sINDELs by removing events seen in sequencing data of a large panel of normal samples. Overall, this filtering removed -35% of all candidate somatic mutations, mostly ones with very low allelic fraction. In order to ensure that no candidate driver mutation were mistakenly removed by the filter, after completing the MutSig process, all filtered events in candidate CLL genes were also manually reviewed using the integrated genome viewer (IGV)!3. In addition all mutations in candidate CLL genes were confirmed by manual inspection as well. The Oncotator tool was used to annotate mutations14. Conservation across 46 vertebrate species was performed and scored as previously described9. Sample contamination by DNA originating from a different individual was assessed using ContEst l5. Median contamination value was 0.1% [inter-quartile range 0.1- 0.3%]. Ig loci mutations were not included in this analysis,
[0094] In the 59 longitudinal samples, Applicants utilized "forced calling" to quantify the number of alternate and reference alleles at sites with somatic mutation detected in a different
sample from the same patient which was taken at a different time point, using the Sam tools suite. Reads were considered if they were not marked as duplicate reads, had a base quality score at the site of interest > 20 and alignment quality score > 5, based on GATK BQSR base quality recalibration11.
[0095] Estimation of and correction for tumor in normal content: For samples from the CLL8 trial, the majority of matched germline DNA samples were obtained from the CD Infraction of PBMC. Applicants found that some of these germline samples contained substantial proportions of tumor DNA, which could significantly decrease the ability to detect somatic mutations with high sensitivity using MuTect. Applicants thus applied deTiN (manuscript in preparation, Taylor-Weiner et a!,), a method for estimating the level of tumor cells in the normal paired sample and recovering somatic mutations that would otherwise be filtered out due to evidence of the mutation in the normal. In brief, Applicants estimate the level of tumor-in- normal (TiN) using two complementary approaches: (i) for tumors with a sufficient number of somatic mutations (total number of sSNVs and sFNDELs >5), Applicants use a linear fit of the respective tumor and normal allele fractions of candidate somatic mutations, and (ii) for tumors with sufficiently large sCNVs, Applicants fit the allele frequency shift of germline heterozygous SNPs to a mixture of tumor and nonnal which provides an independent estimate for TiN. Next Applicants use the TiN estimate to recover sSNVs and sINDELs that are at least a 1000 times more likely to be a somatic mutation than a germline event.
[0096] Significance analysis for recurrently mutated genes: Applicants used MutSig2CV16 to detect candidate cancer genes using three signals of positive selection: (i) increased mutation burden as compared to a background model; (ii) clustering of mutations along the gene; and (iii) enrichment of mutations at likely functional sites.
[0097] Genome-wide copy number analysis: Genome-wide copy number profiles of the CLL samples and their patient-matched germline DNA were estimated directly from the WES data, based on the ratio of CLL sample read-depth to the average read-depth observed in normal samples for that region. Applicants observed a high level of agreement between sCNV detection by exome and standard FISH cytogenetics, with the exception of smaller deletions in the region of chromosome 13ql 4, where 14.5% of cases were missed by WES {Supplementary Table 5). Allelic copy number analysis was then performed by examination of alternate and reference read counts at heterozygous SNP positions (as determined by analysis of the matched normal sample).
These counts were used to infer the contribution of the two homologous chromosomes to the observed copy-ratio in each segment. Further analysis of change-points in these ailelic-ratios was performed using PSCBS17, refining the segmentation. Finally, for each segment, Applicants combined the copy-ratio and allelic data to derive allelic copy-ratios.
[0098] Significant recurrent chromosomal abnormalities were identified using the GISTIC2.0 algorithm18 (v87). Regions with germline copy number variants were excluded from the analysis. Arm level and focal deletion and amplifications filtered by FDR Q<0.01 for significance. Applicants identified 8 recurrent somatic arm level events in 157 of 538 patients (Fig. 1). GISTIC2.0|l analysis yielded 4 significant arm level amplifications, including the previously described amplification of chromosome 8q (n=15) and trisomies of chromosomes 12 (n:==72), 2p in 47 ), and 19 in 6) '!i ' \ Applicants also identified a significant focal amplification peak at 2pl5 with 16 genes, which includes the CLL driver gene XPOl. Applicants noted that XPOl mutations and amplifications involving the 2p arm were mutually exclusive in our cohort. Recurrent arm level deletions included deifllp) (n=34), del(%p) (n 10} , <ie/(18p) (n=13) and ¾/(20p) (n::=7) as previously described20'"'2. Focal deletions included the canonical fife/(13ql4.2) (n=255, containing mir-15a and mir-16-1), del(l lq22.3) (n=118, containing ATM), a large deletion in 6q21 (n=18) (with a peak region spanning 72MB)23, as well as expected deletions in the immunoglobulin (Ig) loci.
[0099] RNA sequencing and data analysis: RNA sequencing (RNAseq) was performed as previously described24. In addition, previously published RNAseq data for additional CLL RNA samples were downloaded with permission from the European Genome-Phenome Archive25, and processed in an identical fashion to the in-house produced libraries. In total, matching WES and RNAseq data were available for 156 samples including 103 samples collected at the DFCI and 53 samples collected by the ICGC. RNAseq BAMs were aligned to the hg!9 genome using the TopHat suite. Each somatic base substitution detected by WES was compared to reads at the same location in RNAseq. Based on the number of alternate and reference reads, a power calculation was obtained with beta-binomial distribution (power threshold used was greater than 90%). A mutation call was deemed negative if no alternate allele reads were observed in RNASeq at the site, as long as RN Aseq was powered to detect an event at the specified location. Differential gene expression analysis was performed for the novel two driver gene mutations which affect at least 5 samples with matched RNAseq and WES data (RPSJ5 [3 DFCI samples
and 2 ICGC samples] and MGA [4 DFCI samples and 2 ICGC samples). Gene expression in transcripts per million (TPM) was quantified using the RSEM algorithm26 (v 1.2.19). Significant batch effects were seen between the DFCI and ICGC samples and were addressed as described herein. After filtering non-expressed genes, gene expression was compared between samples with mutations in one of the studied driver genes and samples that are wild type for this gene. To address the significant batch affected, a generalized linear model (glm) was applied that includes both the mutation status and the batch information; and the P values for the mutation status coefficients were subjected to FDR correction (Q<0.1). Gene set enrichment analysis was performed using the GSEA software"' 7 (V2.2.0), with the pre-ranked list option. Genes were ranked based on the log transformed P values of the glm mutation coefficients.
[00100] Co-occurrence analysis: First, Applicants considered two potential important confounders: prior therapy and IGIiV mutation status, which may affect the proportion of patients affected by specific drivers (Extended Data Fig. 6A-B) resulting in spurious instances of significant low or high co-occurrence. Indeed, despite similar average numbers of coding mutations per sample (24.3 +/- 11.2 versus 23,0 +/- 13.4 in IGIiV mutated vs. unmutated samples, =0.246), the median number of driver mutations per sample was higher in the unmutated IGIIV subtype (3 [IQR 2-4] vs. 1 driver per sample [IQR 1-2], rank sum ><0.00001, note that these do not include IGHV as a driver, as IGHV mutations may represent a physiologic process in B cells). Similarly, compared to treatment-nai've cases, prior exposure to treatment was associated with an increased average number of coding sSNVs and slNDELs (34.9 +/- 22.7 vs. 23.0 +/- 1 1.4, P = 1.16 x 10"'), and a higher median number of drivers (4 vs. 2, P = 3.55 x 10" 9), Of note, to examine the explanation that higher number of mutations simply reflects a longer time from diagnosis to sampling of patients with prior therapy, linear regression model analysis to evaluate the impact of the time between diagnosis and sample acquisition on the association between prior therapy and the number of mutations and drivers was performed. Applicants found that in a model that included both time from diagnosis to sampling and prior therapy, only prior therapy retained significance in terms of a positive association with the number of mutations (P = 0,27147 and =0.00634, respectively) and in terms of a positive association with the number of drivers (P zzz: 0.201 and Pzzz 1.66e-10, respectively). Therefore, to address the effect of IGHV and prior therapy status, significant low or high co-occurrence patterns were retained if the
combined P value28 for the tests in the two subsets after multi-hypotheses correction was significant at Q< .1.
[00101] Estimation of mutation cancer cell fraction using ABSOLUTE: Applicants used the ABSOLUTE algorithm (vl . l.) to calculate the purity, ploidy, and absolute DNA copy- numbers of each sample29, as previously described9. sCNVs were classified as clonal if the modal CCF estimate exceeded 0.85. Modifications were made to the algorithm for the purpose of assessing the clonaiity of sSNVs and sINDELs as follows. For each mutation, the CCF probability density is estimated based on the mutation reference and alternate allele counts (t_ref_count and t_alt_count, respectively), the tumor purity, and local copy number for each homologous allele. The first step is to calculate the allele fraction probability distribution for the tumor alone, excluding the contribution from the normal fraction of cells. The proportion of tumor DNA at a site is:
Tumor DNA fraction -(purity · CNT)/( purity · CNT + CN^ (I -purity))
Where CNT is the local copy number in the tumor cell and CNN is the local copy number in the normal cells (2 in the autosome, and 1 or 2 on the X chromosome depending on gender). The allele fraction probability density in the tumor is estimated from the binomial probability density "binopdf over the range of reference allele counts in the tumor "t ref between 0 and t_ref_count:
w(t ref) hiiiofkifii ref i alt count ref count +t alt count, Tumor DNA fraction) [00102] The probability distribution for the allele fraction in the tumor is then:
p(AFT) =∑t reffwft ref) · betapdffAFr alt count+l,i <_ref+l))/Z
[00103] Where AFT is the full range of allele fractions from 0 to 1 and Z is set such that p(AFY) is normalized to 1.
[00104] At this point, ρ(Α17τ) no longer contains the normal cell component, which simplifies the remaining steps to estimate the mutation cancer cell fraction (CCF). The CCF estimate is integrated over all possible mutation multiplicities "m", the number of mutations per tumor cell. The multiplicity m can range from 1 to q hat I or q hat2 (the local somatic absolute copy numbers of each homologous allele in the tumor). Applicants assume that the mutation occurred before or after any local copy number change and each possible multiplicity is given equal weight wm such that the w's are normalized to 1. AFT is transformed to CCF coordinates by CCF- AFfCNj/m and the probability density for CCF's ranging from 0 to 1 :
p(CCF) -∑m (wsi *p(AFT)) foi- AFr* CNT/m < 1 and iotAFr* CNt/m > 1, the probability density was accumulated at C F=/: piCCF^l) =∑m (wm * p(AFr'CNv'm)) for AFj'CNFm >--- 1
[00105] Clonal mutations were defined as sSNVs or sINDELs with p(CC7' 0,85)>0.5 (ie. median CCF. greater than 0.85).
[00106] Clustering analysis of sSNVs and sINDELs ie 59 CLL sample pairs: Applicants performed WES on matched samples collected at the time of first progression following therapy from 59 of 278 CLL8 subjects (Supplementary Table 9). The median time to progression was 35.1 months (range 5.9-75.5), with relapse samples collected at a median of 7.6 months following documented progression, ail before receipt of subsequent therapy. The two time point CCF clustering procedure was performed as previously described9. Clonal evolution between pretreatment and relapse samples was defined based on the presence of mutations with a P{ CCF > 0.1)>0.5. Branched evolution was classified when a dominant clone in the pre-treatment sample was replaced by sibling dominant clone. This pattern was indicated by a CCF decrease of the mutations in the pre-treatment dominant clone co-occurring with a CCF increase of mutations in the relapse dominant clone. In contrast, linear evolution (replacement of a parent clone by its progeny) was indicated when the increase in CCF of the relapse dominant clone was not ccompanied by a decrease in CCF of the pre-treatment dominant clone,
[00107] For the CLL driver analysis (Fig. SO), a significant change in CCF over time (red or blue) was determined if the 95% CIs of the CCF in the pre-treatment and relapse sample did not overlap. For each driver, a binomial test was performed to assess whether the proportion of instances within each category (increases, decreases, stable) significantly exceeded 0.5 (with a BH FDR correction, QO. l).
[00108] Deep sequencing of somatic single nucleotide variants Targeted deep sequencing was performed using microfluidic PGR (Access Array System, Fluidigm). Six unmatched saliva samples were included in this analysis to assist with the quantification of background sequencing error noise. Target-specific primers were designed to flank sites of interest and produce amplicons of 200 bp ± 20 bp. Per well, molecularly barcoded, lilumina-compatible specific
oligonucleotides containing sequences complementary to the primer tails were added to the Fluidigm Access Array chip together with genomic DNA samples (20-50 ng of input) such that all amplicons for a given DNA sample shared the same index, and PGR was performed according to the manufacturer's instructions. From each individual collection well from the Fluidigm chip, indexed libraries were recovered for each sample, quantified using picogreen, and then normalized for uniformity across libraries. Resulting normalized libraries were loaded on the MiSeq instrument and sequenced using paired-end 150 bp sequencing reads30. Applicants confinned the presence of a mutation in a sample if the fraction of alternate reads exceeded that in the normal control samples (beta binomial test, FDR Q<0.1).
[00109] Order of mutations analysis: To investigate the question of temporal ordering of driver appearance in CLL, the driver clonality patterns in the 501 of 538 samples which were treatment na'ive were studied using the following approach: Whenever a driver event dl is clonal and another driver event d2 is subclonal in the same sample, this pattern indicates that dl was acquired before d2 (denoted by dl →d2), and allow us to draw an edge between these two drivers (out-going for dl, and in-going for d2). Assuming the temporal ordering of a driver pair is random, Applicants can apply hypothesis testing to find significant temporal orderings among drivers in this data set31. Applicants classified driver events as early, late or intermediary/not powered by counting the in-degrees and out-degrees of each driver event across the 501 samples and applying a two-tailed binomial test to quantify whether a driver event has a significantly greater number of out-degrees (early), a significantly greater number of in-degrees (late) or no significant preference (intermediary/not powered). To account for multiple hypothesis testing, Applicants additionally calculate the corresponding q-values as a measure of significance in terms of the false discovery rate32.
[00110] To infer a temporal order between any two pairs of drivers, all known CLL driver pairs were considered for which at least 5 clonal -subclonal orderings were observed, as this was the minimal number of observations powered for statistical significance (P<0.1) to detect a completel unidirectional relationship (i.e., d__l is clonal and d_2 is subclonal in all five pairing). Two-tailed binomial tests were used to quantify the confidence in the temporal ordering of each pair of driver mutations. To account for multiple hypothesis testing, the corresponding q-values were further calculated as a measure of significance in terms of the false discovery rate32.
[00111] Statistical methods Statistical analysis was performed with MATLAB (MathWorks, Natiek, MA), R version 2.1 1.1 and SPSS version 21 (IBM, NYC, NY). Categorical variables were compared using the Pearson Chi-square test or Fisher Exact test as appropriate, and continuous variables were compared using non-parametric rank-sum tests. Statistical analyses of data from the CLL8 clinical trial were performed on an intention-to-treat basis meaning that all eligible subjects with available samples were analyzed as randomized. Time to event analyses were done for progression free survival (PFS), which was defined as the time from randomization to disease progression or death, and for overall survival (OS), which was understood as the time between randomization and death. Time to event data were estimated by the Kaplan-Meier method, and differences between groups were assessed using two-sided non- stratified log-rank tests. Additionally, hazard ratios (HR) and 95% confidence intervals (CI) were calculated using unadjusted and adjusted Cox regression modeling. Independent factors for PFS and OS were identified by multivariable analysis using Cox proportional hazards regression models.
Materials References
1 Hailek, M. et al. Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: a randomised, open-label, phase 3 trial. Lancet 376, 1164- 1174, doi : 10.1016/SO 140-6736( 10)61381 -5 (2010) ,
2 Rassenti, L. et al. Relative value of ZAP-70, CD38, and immunoglobulin mutation status in predicting aggressive disease in chronic lymphocytic leukemia. Blood 112, 1923-1930, doi:blood-2007-05-092882 [pii] 10.1 182/blood-2007-05-092882 (2008).
3 Smoley, S. A. et al Standardization of fluorescence in situ hybridization studies on chronic lymphocytic leukemia (CLL) blood and marrow cells by the CLL Research Consortium. Cancer Genet Cytogenet 203, 141-148, doi: 10.1016/j .cancergencyto.2010.08.009 (2010).
4 Dohner, H. et al. Genomic aberrations and survival in chronic lymphocytic leukemia. N Engl J Med 343, 1910-1916, doi:MJBA-432602 [pii] 10.1056/NEJM200012283432602
(2000).
5 Chapman, M. A. et al. Initial genome sequencing and analysis of multiple myeloma. Nature 471, 467-472, doi: 10.1038/nature09837 (201 1 ).
6 Berger, M. F. et al The genomic complexity of primary human prostate cancer. Nature
470, 214-220, doi: 10.1038/nature09744 (2011).
7 Fisher, S. et al. A scalable, fully automated process for construction of sequence-ready human exome targeted capture libraries. Genome Biol 12, Rl, doi: 10.1186/gb-201 1-12-1- rl (2011).
8 Stilgenbauer, S. et al Gene mutations and treatment outcome in chronic lymphocytic leukemia: results from the CLL8 trial. Blood 123, 3247-3254, doi: 10.1182/biood-2014- 01-546150 (2014).
9 Landau, D, A. et al. Eivoiution and impact of subclonal mutations in chronic lymphocytic leukemia. Cell 152, 714-726, doi : 10.1016/j . cell .2013.01.019 (2013).
10 Quesada, V. et al. Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia. Nat Genet 44, 47-52, doi: 10.1038/ng. l032
(2012) .
11 DePristo, M. A. et al. A framework for variation discovery and genotyping using nextgeneration DNA sequencing data. Nat Genet 43, 491-498, doi: 10.1038/ng.806 (201 1).
12 Cibulskis, K. et al Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nature Biotechnol 31, 213-219, doi : 10.1038/nbt.2514
(2013) .
13 Robinson, J. T. et al Integrative genomics viewer. Nature Biotechnol 29, 24-26, doi : 10.1038/nbt.1754 (201 1).
14 Ramos, A. H. et al Oncotator: Cancer Variant Annotation Tool. Hum Mutat, doi : 10, 1002/humu,22771 (2015).
15 Cibulskis, K. et al ContEst: estimating cross-contamination of human samples in nextgeneration sequencing data. Bioinformatics 27, 2601-2602, doi: 10.1093/bioinformatics/¾ir446 (2011).
16 Lawrence, M. S. et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 505, 495-501 , doi: 10.1038/naturel2912 (2014).
17 Olshen, A. B. et al. Parent-specific copy number in paired tumor-normal studies using circular binary segmentation. Bio informatics 27, 2038-2046, doi: 10.1093/bioinformatics btr329 (201 ).
18 Mermel, C. H. et al. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol 12, R41 , doi: 10.1186/gb-201 l-12-4-r41 (2011).
19 Mermel, C. H. et al. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol 12, R41, doi : 10. 186/gb-2011-12-4-r41 (201 ).
20 Edelmann, J. et al. High-resolution genomic profiling of chronic lymphocytic leukemia reveals new recurrent genomic alterations. Blood 120, 4783-4794, doi : 10.1 182/biood- 2012-04- 423517 (2012).
21 Seilmann, L. et al. Trisomy 19 is associated with trisomy 12 and mutated IGHV genes in B-chronic lymphocytic leukaemia. Br J Haematol 138, 217-220, doi :BJH6636 [pii] 10.11 1 1/j .1365-2141.2007.06636.x (2007).
22 Rudenko, H. el al. Characterising the TP53-deleted subgroup of chronic lymphocytic leukemia: an analysis of additional cytogenetic abnormalities detected by interphase fluorescence in situ hybridisation and array-based comparative genomic hybridisation. Leak Lymphoma 49, 1879-1886, doi :904754482 [pii] 10, 1080/10428190802345902 (2008).
23 Cuneo, A. et al. Chronic lymphocytic leukemia with 6q- shows distinct hematological features and intermediate prognosis. Leukemia 18, 476-483, doi: 10.1038/sj . leu.2403242 (2004).
24 Landau, D. A. et al. Locally disordered methylation forms the basis of intratumor methylome variation in chronic lymphocytic leukemia. Cancer Cell 26, 813-825, doi: 10.1016/j .ceeli.2014.10.012 (2014).
25 Ferreira, P. G. et al. Transcriptome characterization by RNA sequencing identifies a major molecular and clinical subdivision in chronic lymphocytic leukemia. Genome Res 24, 212- 226, doi : 10.1101 /gr.152132.1 12 (2014).
26 Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC hioinformatics 12, 323, doi: 10.1186/1471 - 2105-12- 323 (2011).
27 Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. PNAS 102, 15545-15550, doi: 10.1073/pnas.0506580102 (2005),
28 Whitlock, M. C. Combining probability from independent tests: the weighted Z-method is superior to Fisher's approach. J Evol Biol 18, 1368-1373, doi: 10.1 111/j .1420- 9101.2005,00917.x (2005),
29 Carter, S. L. et al Absolute quantification of somatic DNA alterations in human cancer. Nature Biotechnol 30, 413-421, doi: 10.1038/nbt.2203 (2012).
30 Lohr, j. G. et al. Discover}' and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing. PNAS 109, 3879-3884, doi: 10.1073/pnas.1 121343109 [pii] (2012).
31 Wang, J. et al. Tumor evolutionary directed graphs and the history of chronic lymphocytic leukemia, el fe 3, doi: 10.7554/eLife.02869 (2014).
32 Storey, J. D. & Tibshirani, R. Statistical significance for genomewide studies. PNAS 100, 9440-9445, doi : 30, 1073/pnas.3 530509100 (2003).
* * *
[00112] Having thus described in detail preferred embodiments of the present invention, it is to be understood that the invention defined by the above paragraphs is not to be limited to particular details set forth in the above description as many apparent variations thereof are possible without departing from the spirit or scope of the present invention.
Claims
1. A method of treating chronic lymphocytic leukemia (CLL) in a subject in need thereof comprising identifying the presence of gene mutations and somatic copy number variations (CNV) in a genomic DNA sample obtained from the subject, wherein the mutated genes and somatic copy number variations (CNV) comprise del(13q), SF3B1, ATM, del(llq), tril2, TPS 3, NOTCH!, POT I, amp(2p), dei(17p), CHD2, XPOI, RPS15, BIRC3, BRAF, IGLL5, del(8p), MGA, MYD88, del(6q21), EGR2, FBXW7, amp(8q), DDX3X, KRAS, BCOR, 1KZF3, MAP2K1, ZMYM3, IRF4, SAMHD1, BAZ2A, CARD J 1, FUBPl, HIST1H1E, MED12, NRAS, NXFl, del (18p), DYRK1A, PTPNU, del(20p), ELF 4, TRAP 2, XP04, triI9, BRCC3, EWSR1, FAM50A, TRAP 3, ASXL1, CHEK.2, GNB1, HIST1H1B, and PIMJ, wherein the subject is treated if the presence of a mutated gene and/or somatic copy number variation (CNV) is identified.
2. The method according to claim 1, wherein the patient has stage 0 CLL.
3. The method according to claims 1 or 2, wherein the presence of mutations in NRAS, KRAS, BRAF', and/or MAP2K1 indicate treating with an inhibitor of the MAPK-ERK pathway.
4. The method according to claim 3, wherein the mutation in BRAF is not V600E and wherein the treatment includes a MEK inhibitor and not a BRAF' inhibitor.
5. The method according to claims 1 or 2, wherein the presence of mutations in RPS15 indicate an adverse outcome and the subject is treated with an alternative treatment regimen.
6. The method according to claims 1 or 2, wherein the presence of mutations in IKZF3 indicate an adverse outcome and the subject is treated with an alternative treatment regimen.
7. The method according to claim 6, wherein the mutation in IKZP3 is an L162R substitution.
8. The method according to claims 1 or 2, wherein the presence of mutations in deli' !3q) . MYD88, and/or CFLD2 indicate decreased clinical aggressiveness.
9. The method according to claims 1 or 2, wherein the presence of mutations in TP53, SF3B1 and/or XPOI indicate shorter progression-free survival (PFS) and overall survival (OS),
10. The method according to any of claims 1 to 9, wherein the sample is obtained prior to treatment.
11. The method according to any of claims 1 to 10, further comprising determining mutations in /(;/// wherein the presence of unmutated IGHV indicates increased clinical aggressiveness and the presence of mutated IGHV indicates decreased clinical aggressiveness.
12. A method of determining whether a subject having chronic lymphocytic leukemia (CLL) would derive a clinical benefit of early treatment comprising determining the presence of gene mutations and somatic copy number variations (CNV) in a genomic DNA sample obtained from the subject, wherein the mutated genes and somatic copy number variations (CNV) comprise del/13q), SF3B1, A TM, del(llq), in 12, Ί 53, NOTCH 1, POT1, amp(2p), del(17p), CHD2, XPOl, RPS15, BIRC3, BRAF, IGLL5, del(8p), MGA, MYD88, del(6q21), EGR2, FBXW7, amp(8q), DDX3X, KRAS, BCOR, IKZF3, MAP2K1, ZMYM3, IRF4, SAMHDJ, BAZ2A, CARDll, FlJBPl, HISTTHIE, MED 12, NRAS, NXFl, del (18p), DYRK1A, PTPN! I, del(20p), ELF4, TRAF2, XP04, tril9, BRCC3, EWSRI, FAM50A, TRAF3, ASXLI, CHEK2, GNBl, HISTIHIB, and PIMI, wherein the presence of a mutated gene and/or somatic copy number variation (CNV) indicates that the subject would derive a clinical benefit of early treatment.
13. A method of predicting survivability of a subject having chronic lymphocytic leukemia (CLL) comprising determining the presence of gene mutations and somatic copy number variations (CNV) in a genomic DNA sample obtained from the subject, wherein the mutated genes and somatic copy number variations (CNV) comprise<3k/(73¾ , SF3BI, A TM,' del/1 Iq), tril2, TPS 3, NOTCH 1, POT1, amp(2p), del/Up), CHD2, XPOl, RPS15, BIRC3, BMP, IGLL5, del(8p), MGA, MYD88, del(6q21), EGR2, FBXW7, amp(8q), DDX3X, KRAS, BCOR, IKZF3, MAP2K1, ZMYM3, IRF4, SAMHDl, BAZ2A, CARDll, FlJBPl, HISTIHIE, MED12, NRAS, NXFL del (18p), DYRK1A, PTPNll, delQQp), ELF 4, TRAF2, XP04, tril9, BRCC3, EWSRI, FAM50A, TRAF3, ASXLI, CHEK2, GNBl, HISTIHIB, and PIMI, wherein the presence of a mutated gene and/or somatic copy number variation (CNV) indicates that the subject is less likely to survive.
14. A method of identifying a candidate subject for a clinical trial for a treatment protocol for chronic lymphocytic leukemia (CLL) comprising determining the presence of gene mutations and somatic copy number variations (CNV) in a genomic DNA sample obtained from the subject, wherein the mutated genes and somatic copy number variations (CNV) comprise
del(13q), SI- 311 !. AIM., del(llq), trill, TP 53, NOTCH!, POTl, amp(2p), del(17p), CHDl, XPOl, RIJS15, BIRC3, BMP, IGLL5, del(8p), MGA, MYP 88, del(6q21), EGR2, FBXW7, ampfiq), DDX3X, KRAS, BCOR, IKZF3, MAP1K1, ZMYM3, IRF4, SAMHD1, BAZ2A, CARDll, FlJBPl, HIST1H1E, MED 12, NRAS, NXF1, del (I8p), DYRKIA, PTPN11, del(20p), ELF4, TRAF2, XP04, tril9, BRCC3, EWSR1, FAM50A, TRAF3, ASXL1, CHEK2, GNB1, HIST IE IB, and PIML wherein the presence of a mutated gene and/or somatic copy number variation (CNV) indicates that the subject is a candidate for the clinical trial.
15. A method of detecting chronic lymphocytic leukemia (CLL) in a subject comprising determining the presence of gene mutations and somatic copy number variations (CNV) in a genomic DNA sample obtained from the subject, wherein the mutated genes and somatic copy number variations (CNV) compri sodelf I3q), SF3B1, ATM, del(llq), trill, TP 53, NOTCH 1, POTl, amp(2p), del(17p), CHD2, XPOl, RPS15, BIRC3, BRAF, IGLL5, del(8p), MGA, MYD88, del(6q21), EGR2, FBXW7, amp(8q), DDX3X, KRAS, BCOR IKZF3, MAP2K1, ZMYM3, IRF4, SAMHD1, BAZ2A, CARDll, FlJBPl, HISTIHIE, MFD 12. NRAS, NXF1, del (18p), DYRKIA, PTPN11, del (20p), ELF 4, TRAFl, XP04, tril9, BRCC3, EWSRl, FAM50A, TRAF3, ASXIA, CHEK1, GNB1, HIST1H1B, and PIM1, whereby the presence of one or more mutations and/or CNV indicates that the subject has a greater than 90% probability of having CLL.
16. A method of identifying a subject at elevated risk of having CLL with rapid disease progression comprising:
(a) analyzing genomic DNA in a sample obtained from a subject having or suspected of having chronic lymphocytic leukemia (CLL.) for the presence of gene mutations and somatic copy number variations (CNV), wherein the mutated genes and somatic copy number variations (CNV) comynsodel/lSq), SF3B1, AIM, del(llq), iril2, TPS 3, NOTCH I POTl, amp(lp), del(17p), CHDl, XPOl, RPS15, BIRC3, BR iF, IGLL5, del(8p), MGA, MYD88, del(6qll), EGRl, FBXW7, amp(8q), DDX3X, KRAS, BCOR, 1KZF3, MAP IK 1, ZMYM3, 1RF4, SAMHDl, BAZIA, CARDll, FUBPl, HISTIHIE, MED 12, NRAS, NXF1, del (18p), DYRKIA, PTPNll, del(lOp), ELF4, TRAFl, XP04, tril9, BRCC3, EWSR1, FAM50A, TRAF3, ASXL1, CHEK1, GNB1, HIST1H1B, and TI I :
(b) determining whether the mutations and/or somatic copy number variations are clonal or subclonal, and
(c) identifying the subject as a subject at elevated risk of having CLL with rapid disease progression if at least one mutation and/or somatic copy number variation is sub clonal.
17. The method according to claim 16, further comprising treating a subject identified as a subject at elevated risk of having CLL with rapid disease progression.
18. The method according to any one of claims 1 to 17, wherein mutations in more than one risk allele are analyzed.
19. The method according to any one of claims 1 to 18, wherein the method is performed before treatment.
20. The method according to any one of claims 1 to 18, wherein the method is performed after treatment.
21. The method according to any one of claims 1 to 20, further comprising repeating the method every 6 months or if there is a change in clinical status.
22. The method according to any one of claims 1 to 21, wherein the genomic DNA sample is obtained from peripheral blood, bone marrow, or lymph node tissue.
23. The method according to any one of claims 1 to 22, wherein the genomic DNA is analyzed using whole genome sequencing (WGS), whole exome sequencing (WES), single nucleotide polymorphism (SNP) analysis, deep sequencing, targeted gene sequencing, hybridization to an array, or any combination thereof.
24. The method according to any one of claims 16 to 23, wherein clonal or subclonal mutations, CNV's and/or populations of cells are detected using whole genome sequencing (WGS), whole exome sequencing (WES), single nucleotide polymorphism (SNP) analysis, deep sequencing, targeted gene sequencing, hybridization to an array, or any combination thereof.
25. The method according to any one of claims 1-24, wherein the gene mutation is a missense mutation, frameshift indel, inframe indel, splice site mutation, or nonsense mutation.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201562244533P | 2015-10-21 | 2015-10-21 | |
US62/244,533 | 2015-10-21 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2017070497A1 true WO2017070497A1 (en) | 2017-04-27 |
Family
ID=57223793
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2016/058164 WO2017070497A1 (en) | 2015-10-21 | 2016-10-21 | Methods and compositions for use of driver mutations in cll |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2017070497A1 (en) |
Cited By (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN108570503A (en) * | 2018-05-29 | 2018-09-25 | 成都中创清科医学检验所有限公司 | A kind of primer and its detection method for detecting chronic lymphocytic leukemia gene polymorphism sites |
US10364467B2 (en) | 2015-01-13 | 2019-07-30 | The Chinese University Of Hong Kong | Using size and number aberrations in plasma DNA for detecting cancer |
WO2020006431A1 (en) * | 2018-06-29 | 2020-01-02 | Rady Children's Hospital Research Center | Method and system for sample identity assurance |
US10741270B2 (en) | 2012-03-08 | 2020-08-11 | The Chinese University Of Hong Kong | Size-based analysis of cell-free tumor DNA for classifying level of cancer |
DE102020102143B3 (en) * | 2020-01-29 | 2021-03-04 | Cellphenomics GmbH | Method for determining whether treatment of a cancer disease is to be started or continued, a biomarker which corresponds to at least one marker gene, and a use of the biomarker in the method according to the invention |
WO2021178595A3 (en) * | 2020-03-04 | 2021-10-21 | Foundation Medicine, Inc. | Bcor rearrangements and uses thereof |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2013086464A1 (en) * | 2011-12-07 | 2013-06-13 | The Broad Institute, Inc. | Markers associated with chronic lymphocytic leukemia prognosis and progression |
-
2016
- 2016-10-21 WO PCT/US2016/058164 patent/WO2017070497A1/en active Application Filing
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2013086464A1 (en) * | 2011-12-07 | 2013-06-13 | The Broad Institute, Inc. | Markers associated with chronic lymphocytic leukemia prognosis and progression |
Non-Patent Citations (8)
Title |
---|
ANTONIO CUNEO ET AL: "Modern treatment in chronic lymphocytic leukemia: impact on survival and efficacy in high-risk subgroups", CANCER MEDICINE, vol. 3, no. 3, 19 March 2014 (2014-03-19), GB, pages 555 - 564, XP055335723, ISSN: 2045-7634, DOI: 10.1002/cam4.226 * |
D. ROSSI ET AL: "Mutations of the SF3B1 splicing factor in chronic lymphocytic leukemia: association with progression and fludarabine-refractoriness", BLOOD, vol. 118, no. 26, 28 October 2011 (2011-10-28), pages 6904 - 6908, XP055052912, ISSN: 0006-4971, DOI: 10.1182/blood-2011-08-373159 * |
DANIEL L. VAN DYKE ET AL: "A comprehensive evaluation of the prognostic significance of 13q deletions in patients with B-chronic lymphocytic leukaemia", BRITISH JOURNAL OF HAEMATOLOGY, vol. 148, no. 4, 1 February 2010 (2010-02-01), GB, pages 544 - 550, XP055336090, ISSN: 0007-1048, DOI: 10.1111/j.1365-2141.2009.07982.x * |
DEL GIUDICE ILARIA ET AL: "NOTCH1 mutations in +12 chronic lymphocytic leukemia (CLL) confer an unfavorable prognosis, induce a distinctive transcriptional profiling and refine the intermediate prognosis of +12 CLL.", HAEMATOLOGICA MAR 2012, vol. 97, no. 3, March 2012 (2012-03-01), pages 437 - 441, XP002766050, ISSN: 1592-8721 * |
MATTHEW S. DAVIDS ET AL: "Controversial fluorescence in situ hybridization cytogenetic abnormalities in chronic lymphocytic leukaemia: new insights from a large cohort", BRITISH JOURNAL OF HAEMATOLOGY, vol. 170, no. 5, 1 June 2015 (2015-06-01), GB, pages 694 - 703, XP055336086, ISSN: 0007-1048, DOI: 10.1111/bjh.13498 * |
NABHAN CHADI ET AL: "Predicting Prognosis in Chronic Lymphocytic Leukemia in the Contemporary Era.", JAMA ONCOLOGY OCT 2015, vol. 1, no. 7, 7 May 2015 (2015-05-07), pages 965 - 974, XP002766052, ISSN: 2374-2445 * |
PUIGGROS ANNA ET AL: "Genetic Abnormalities in Chronic Lymphocytic Leukemia: Where We Are and Where We Go", BIOMED RESEARCH INTERNATIONAL, 2014, XP002766051 * |
STEVEN J. T. HUANG ET AL: "Influence of clone and deletion size on outcome in chronic lymphocytic leukemia patients with an isolated deletion 13q in a population-based analysis in British Columbia, Canada", GENES CHROMOSOMES & CANCER., vol. 55, no. 1, 22 September 2015 (2015-09-22), US, pages 16 - 24, XP055335697, ISSN: 1045-2257, DOI: 10.1002/gcc.22294 * |
Cited By (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US10741270B2 (en) | 2012-03-08 | 2020-08-11 | The Chinese University Of Hong Kong | Size-based analysis of cell-free tumor DNA for classifying level of cancer |
US11031100B2 (en) | 2012-03-08 | 2021-06-08 | The Chinese University Of Hong Kong | Size-based sequencing analysis of cell-free tumor DNA for classifying level of cancer |
US10364467B2 (en) | 2015-01-13 | 2019-07-30 | The Chinese University Of Hong Kong | Using size and number aberrations in plasma DNA for detecting cancer |
CN108570503A (en) * | 2018-05-29 | 2018-09-25 | 成都中创清科医学检验所有限公司 | A kind of primer and its detection method for detecting chronic lymphocytic leukemia gene polymorphism sites |
WO2020006431A1 (en) * | 2018-06-29 | 2020-01-02 | Rady Children's Hospital Research Center | Method and system for sample identity assurance |
DE102020102143B3 (en) * | 2020-01-29 | 2021-03-04 | Cellphenomics GmbH | Method for determining whether treatment of a cancer disease is to be started or continued, a biomarker which corresponds to at least one marker gene, and a use of the biomarker in the method according to the invention |
WO2021178595A3 (en) * | 2020-03-04 | 2021-10-21 | Foundation Medicine, Inc. | Bcor rearrangements and uses thereof |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Newman et al. | Genomes for kids: the scope of pathogenic mutations in pediatric cancer revealed by comprehensive DNA and RNA sequencing | |
US20220010385A1 (en) | Methods for detecting inactivation of the homologous recombination pathway (brca1/2) in human tumors | |
US11834718B2 (en) | Compositions and methods for diagnosing, evaluating and treating cancer by means of the DNA methylation status | |
JP7573443B2 (en) | Methods for cancer detection and monitoring using personalized detection of circulating tumor dna - Patents.com | |
Quek et al. | Clonal heterogeneity of acute myeloid leukemia treated with the IDH2 inhibitor enasidenib | |
JP6700333B2 (en) | Methods and materials for assessing loss of heterozygosity | |
Shibuya et al. | Identification of somatic genetic alterations in ovarian clear cell carcinoma with next generation sequencing | |
Herling et al. | Complex karyotypes and KRAS and POT1 mutations impact outcome in CLL after chlorambucil-based chemotherapy or chemoimmunotherapy | |
Juskevicius et al. | Distinct genetic evolution patterns of relapsing diffuse large B-cell lymphoma revealed by genome-wide copy number aberration and targeted sequencing analysis | |
Saddler et al. | Comprehensive biomarker and genomic analysis identifies p53 status as the major determinant of response to MDM2 inhibitors in chronic lymphocytic leukemia | |
Dolnik et al. | Commonly altered genomic regions in acute myeloid leukemia are enriched for somatic mutations involved in chromatin remodeling and splicing | |
Agathangelidis et al. | Highly similar genomic landscapes in monoclonal B-cell lymphocytosis and ultra-stable chronic lymphocytic leukemia with low frequency of driver mutations | |
WO2017070497A1 (en) | Methods and compositions for use of driver mutations in cll | |
Smith et al. | Candidate DNA repair susceptibility genes identified by exome sequencing in high-risk pancreatic cancer | |
Bolli et al. | Next-generation sequencing for clinical management of multiple myeloma: ready for prime time? | |
Brecqueville et al. | Array comparative genomic hybridization and sequencing of 23 genes in 80 patients with myelofibrosis at chronic or acute phase | |
WO2012106559A1 (en) | Biomarkers and methods of use thereof | |
US9982304B2 (en) | ARID1A and PPP2R1A mutations in cancer | |
AU2022275430A1 (en) | Genetic abnormalities in plasma cell dyscrasias | |
WO2015112619A1 (en) | Methods and systems for detecting genetic mutations | |
Degan et al. | Analysis of IgVH gene mutations in B cell chronic lymphocytic leukaemia according to antigen‐driven selection identifies subgroups with different prognosis and usage of the canonical somatic hypermutation machinery | |
Meazza et al. | AKT 1 and BRAF mutations in pediatric aggressive fibromatosis | |
Bloehdorn et al. | Multi-platform profiling characterizes molecular subgroups and resistance networks in chronic lymphocytic leukemia | |
Nagano et al. | Comprehensive molecular profiling of pulmonary pleomorphic carcinoma | |
Park et al. | Distribution of somatic mutations of cancer-related genes according to microsatellite instability status in Korean gastric cancer |
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: 16790502 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 16790502 Country of ref document: EP Kind code of ref document: A1 |