Abstract

Background:

BRCA germline mutations are being targeted for development of PARP inhibitors. BRCA genes collaborate with several others in the Fanconi Anemia (FA) pathway. We screened cancer patients’ tumors for FA functional defects then aimed to establish the safety/feasibility of administering PARP inhibitors as monotherapy and combined with a DNA-breaking agent.

Methods:

Patients underwent FA functional screening for the presence (or lack) of tumor FancD2 nuclear foci formation on their archival tumor material, utilizing a newly developed method (Fanconi Anemia triple-stain immunofluorescence [FATSI]), performed in a Clinical Laboratory Improvement Amendments–certified laboratory. FATSI-negative patients were selected for enrollment in a two-arm dose escalation trial of veliparib, or veliparib/mitomycin-C (MMC).

Results:

One hundred eighty-five of 643 (28.7%) screened patients were FATSI-negative. Sixty-one received veliparib or veliparib/MMC through 14 dose levels. Moderate/severe toxicities included fatigue (DLT at veliparib 400mg BID), diarrhea, and thrombocytopenia. Recommended doses are 300mg BID veliparib and veliparib 200mg BID for 21 days following 10mg/m 2 MMC every 28 days. Six antitumor responses occurred, five in the combination arm (3 breast, 1 ovarian, 1 endometrial [uterine], and 1 non–small cell lung cancer). Two patients have received 36 and 60 cycles to date. BRCA germline analysis among 51 patients revealed five deleterious mutations while a targeted FA sequencing gene panel showed missense/nonsense mutations in 29 of 49 FATSI-negative tumor specimens.

Conclusions:

FATSI screening showed that a substantial number of patients’ tumors have FA functional deficiency, which led to germline alterations in several patients’ tumors. Veliparib alone or with MMC was safely administered to these patients and produced clinical benefit in some. However, a better understanding of resistance mechanisms in this setting is needed.

Mutations of the breast cancer susceptibility (BRCA) genes have been identified as potential predictors of antitumor response to PARP inhibitors ( 1–5 ). They collaborate with several others in the Fanconi Anemia (FA) repair pathway ( 6–19 ). FA patients have a high incidence of malignancies and their cells exhibit hypersensitivity to DNA cross-linking agents such as mitomycin C (MMC) and cisplatin ( 20–22 ). FA falls into 17 complementation group subtypes ( 12–19 ). Eight of these proteins and three associated factors are subunits of an FA core complex, a nuclear E3 ubiquitin ligase ( 12–14 , 22 ). Monoubiquitination of FancD2 and FancI by the FA core complex followed by nuclear colocalization with other DNA damage response proteins results in nuclear foci of repair ( Figure 1 ) ( 15 ). Any alteration that disrupts components of the core complex abrogates its E3 ligase function, leading to defective mono-ubiquitination and no repair foci formation ( 7 , 22 ).

The Fanconi Anemia (FA) pathway and formation of repair foci. Following DNA interstrand crosslink damage, the FANCM-FAAP24-MHF1-MHF2 anchor complex recruits the FA core complex I, which functions to activate FANCD2 and FANCI by mono-ubiquitinating the proteins. The activated FANCD2 and FANCI heterodimers are subsequently transported to subnuclear foci, which in collaboration with additional genes result in homologous recombination DNA repair.
Figure 1.

The Fanconi Anemia (FA) pathway and formation of repair foci. Following DNA interstrand crosslink damage, the FANCM-FAAP24-MHF1-MHF2 anchor complex recruits the FA core complex I, which functions to activate FANCD2 and FANCI by mono-ubiquitinating the proteins. The activated FANCD2 and FANCI heterodimers are subsequently transported to subnuclear foci, which in collaboration with additional genes result in homologous recombination DNA repair.

Disruptions of the FA/BRCA cascade have been noted in sporadic cancers, including epigenetic silencing of the FA core complex, mutations of FA/BRCA genes, or modification of encoded products ( 23–25 ). Cancers with a defective FA/BRCA pathway are likely to be more sensitive to cross-link-based therapy, and treatments in which an additional repair mechanism is targeted may have antitumor activity or provide therapy sensitization ( 26–31 , 57 ).

We hypothesized that given the number of modifications that could interfere with FA pathway functionality a substantial number of patients would be good candidates for PARP inhibitor or cross-link cytotoxic-based therapy. To identify these patients, we developed an assay, FancD2/DAPI/Ki67 (Fanconi Anemia triple-stain immunofluorescence [FATSI]), which permits the observation (or lack thereof) of FancD2 foci formation in proliferating cells ( 32 ). The FATSI assay demonstrated reliable performance in paraffin-embedded (FFPE) archival tumor material and underwent validation in a Clinical Laboratory Improvement Amendments (CLIA)–certified laboratory, thus it is suitable for large-scale screening.

In this first of its kind trial we set out to: 1) screen cancer patients to identify those with FA functional defects in their tumors, 2) establish the safety/feasibility of PARP inhibition as monotherapy and in combination with a DNA-breaking agent in these patients, and 3) recommend appropriate doses for subsequent studies.

Methods

Patients

The Institutional Review Boards of The Ohio State University (OSU) and the Georgetown University approved this study (clinicaltrials.gov; NCT01017640). The study was performed in two parts. Patients older than age 18 years with advanced solid malignancies consented to have their existing FFPE tumor tissue screened for FA deficiency by the FATSI assay. Those determined to be FA functionally deficient (FATSI-negative) were offered a place in the therapeutic portion of the trial. Separate written consents for the screening and therapeutic intervention were obtained ( Figure 2 ).

Patient screening and flow diagram. CLIA = Clinical Laboratory Improvement Amendments.
Figure 2.

Patient screening and flow diagram. CLIA = Clinical Laboratory Improvement Amendments.

Other eligibility requirements for the therapeutic portion included progressive disease, less than three previous cytotoxic chemotherapy regimens for metastatic disease, and a lapse of four weeks from chemotherapy or radiation therapy. Prior MMC restricted to topical applications or chemo-embolization was allowed. We required patients have an ECOG performance status of less than 2 and normal organ and marrow function (absolute neutrophil count ≥ 1.5 x 10 9 ; platelets ≥ 100 x 10 9 ; hemoglobin ≥ 9g/dL; serum creatinine and bilirubin ≤ 1.5 x the upper limit of normal; AST/ALT ≤2.5 x the upper limit of normal). Patients were excluded because of pregnancy, active brain metastases, recent history of seizures, uncontrolled concurrent illness, combination antiretroviral therapy, and previous treatment with PARP inhibitors.

Treatment Plan

Patients were allocated to one of two arms, and a 3+3 dose escalation design was followed. On arm 1, patients received the oral PARP inhibitor veliparib as monotherapy, and on arm 2 patients received veliparib combined with MMC. MMC was chosen because of its well-defined role in producing double-strand DNA breaks, its approval for use in a vast number of solid malignancies, demonstration in our laboratory of stimulation of the FA pathway, and because our previous experience has shown it induces sensitivity to veliparib ( 33 ). No pharmacokinetic interactions between veliparib or MMC were anticipated.

To allocate patients to the treatment arms, we used a “Ping-Pong” approach. As 3+3 phase I trials are designed to enroll three patients and evaluate toxicity before evaluating the next dose level or expanding, there is a gap of a few weeks for enrollment. We initiated with the monotherapy arm, continuing to the combination arm once the monotherapy arm at a particular dose level was full. The “Ping-Pong” approach discouraged bias from the referring physician (or patient) for a particular arm (with chemo vs without chemo).

The velaparib starting dose was 50mg twice daily (BID), which had demonstrated PARP catalytic activity inhibition in tumor in a previous report ( 34 ). As a safety precaution for the combination arm, an increase on the duration of administration of 50mg BID veliparib for the first three patients cohorts (7, 14, and 21 days, every 28 days) following MMC 10mg/m 2 was evaluated prior to escalating veliparib. MMC was administered every 28 days, with a cumulative dose cap of 40mg/m 2 . Thus, no patient in the combination arm would receive a higher dose than a dose previously cleared on the monotherapy arm. Patients experiencing clinical benefit continued on single-agent veliparib once the MMC dose cap was reached.

Dose-limiting toxicity (DLT) was defined as grade 4 neutropenia for more than seven days or with sepsis or fever, grade 4 thrombocytopenia, grade 3–4 nonhematologic toxicity that caused an interruption of veliparib dosing for seven or more days, or any grade 1–2 treatment-related toxicity requiring dose delays for more than four weeks. At least two cycles for DLT evaluation were required in the combination arm whereas one cycle (4 weeks) for the monotherapy arm was considered sufficient. The maximum tolerated dose (MTD) was the dose at which no more than one of six patients experienced DLT. Toxicities were graded according to National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events version 4. Measurable disease was not required. Imaging was repeated every two cycles.

Biomarker and Correlative Studies

Tumor Tissue Screening

Archival tumor tissue was retrieved and sent to the Department of Pathology. Tissue sections were cut to 4 microns, and FATSI staining and interpretation was performed in the CLIA-certified OSUCCC Molecular Pathology Core Laboratory (MPCL) by the same pathologist, as previously described ( 32 ).

BRCA Germline Mutational Analysis

Blood samples (8mL) from patients in the therapeutic portion (receiving veliparib or veliparib/MMC) were collected prior to treatment using blood collection specimen kits obtained from Myriad Genetics (Salt Lake City, Utah), where sequencing for detection of BRCA1 and 2 mutations and the five most common BRCA1 large rearrangements was performed.

Gamma-H2AX and PARP Activity

Blood samples prior to dosing and at two, four, six, and 24 hours on day 1 of cycles 1 and 2 were obtained. PBMC were isolated with BD Vacutainer Cell Preparation Tubes (BD Diagnostics, Franklin Lakes, NJ) and fixed by immersing the cells in 2% formaldehyde solution at 37°C for 20 minutes and then stored in 70% ethanol until batch-analyzed. Quantitative staining for gammaH2AX and PARP activity using enzyme-linked immunosorbent assay was performed at the National Clinical Target Validation Laboratory, NCI Frederick, as previously described ( 35 ).

Repair Sequencing Panel

To evaluate potential genetic alterations that led to the FA functional deficiency, we developed a targeted FA sequencing panel (25 genes) using Agilent’s Haloplex Custom Library system to test a group of random FATSI-negative samples from the screening portion of the trial ( Supplementary Table 1 , available online). The designed panel includes the entire coding sequence of each gene. One cell line and a patient sample with known mutations in FANCC and FANCD2 were positive controls. A library was prepared for each patient specimen as previously described ( 36 ) and sequenced on an Illumina HighSeq2000 to an average coverage of 1000X. Sequencing data was analyzed using the Agilent SureCall software. Variant results were filtered based on read depth and quality, nonsynonymous amino acid changes or splice site base changes, and allelic frequency in the general population.

Results

Screening

From October 2009 to June 2014, 724 patients with solid malignancies consented to FATSI screening. Of these, 71 had insufficient archived tissue, four failed screening because of technical issues, and six either withdrew or died before testing was conducted. FATSI was performed in 643 patients ( Table 1 ). For patients with archived material at OSU, the retrieval of and specimen selection, cutting, and processing took an average of two days. Staining and interpretation at the OSU-MPCL were conducted within 48 hours.

Table 1.

Tissue screening results

Primary tumor typeFATSI negativeFATSI positivePatients tested% FATSI negative
Colon6310416737.7
Rectal591435.7
Breast4110114228.8
Lung non–small cell19789719.6
Lung small cell6253119.4
Ovarian6202623.1
Biliary track8132138.1
Pancreatic3172015.0
Bladder5131827.8
Head & neck6121833.3
Endometrium112137.7
Prostate112137.7
Adenoca unknown primary281020.0
Neuroendocrine36933.3
Mesothelioma25728.6
Cervical0550
Thymic23540.0
Sarcoma12333.3
Melanoma21366.7
Gastro-intestinal stromal tumor12333.3
Stomach12333.3
Small bowell21366.7
Appendix11250.0
Renal11250.0
Adrenal11250.0
Testicular101100.0
Vulva0110
Penile0110
Fallopian0110
Peritoneal101100.0
Hepatic0110
Total18545864328.7
Primary tumor typeFATSI negativeFATSI positivePatients tested% FATSI negative
Colon6310416737.7
Rectal591435.7
Breast4110114228.8
Lung non–small cell19789719.6
Lung small cell6253119.4
Ovarian6202623.1
Biliary track8132138.1
Pancreatic3172015.0
Bladder5131827.8
Head & neck6121833.3
Endometrium112137.7
Prostate112137.7
Adenoca unknown primary281020.0
Neuroendocrine36933.3
Mesothelioma25728.6
Cervical0550
Thymic23540.0
Sarcoma12333.3
Melanoma21366.7
Gastro-intestinal stromal tumor12333.3
Stomach12333.3
Small bowell21366.7
Appendix11250.0
Renal11250.0
Adrenal11250.0
Testicular101100.0
Vulva0110
Penile0110
Fallopian0110
Peritoneal101100.0
Hepatic0110
Total18545864328.7
Table 1.

Tissue screening results

Primary tumor typeFATSI negativeFATSI positivePatients tested% FATSI negative
Colon6310416737.7
Rectal591435.7
Breast4110114228.8
Lung non–small cell19789719.6
Lung small cell6253119.4
Ovarian6202623.1
Biliary track8132138.1
Pancreatic3172015.0
Bladder5131827.8
Head & neck6121833.3
Endometrium112137.7
Prostate112137.7
Adenoca unknown primary281020.0
Neuroendocrine36933.3
Mesothelioma25728.6
Cervical0550
Thymic23540.0
Sarcoma12333.3
Melanoma21366.7
Gastro-intestinal stromal tumor12333.3
Stomach12333.3
Small bowell21366.7
Appendix11250.0
Renal11250.0
Adrenal11250.0
Testicular101100.0
Vulva0110
Penile0110
Fallopian0110
Peritoneal101100.0
Hepatic0110
Total18545864328.7
Primary tumor typeFATSI negativeFATSI positivePatients tested% FATSI negative
Colon6310416737.7
Rectal591435.7
Breast4110114228.8
Lung non–small cell19789719.6
Lung small cell6253119.4
Ovarian6202623.1
Biliary track8132138.1
Pancreatic3172015.0
Bladder5131827.8
Head & neck6121833.3
Endometrium112137.7
Prostate112137.7
Adenoca unknown primary281020.0
Neuroendocrine36933.3
Mesothelioma25728.6
Cervical0550
Thymic23540.0
Sarcoma12333.3
Melanoma21366.7
Gastro-intestinal stromal tumor12333.3
Stomach12333.3
Small bowell21366.7
Appendix11250.0
Renal11250.0
Adrenal11250.0
Testicular101100.0
Vulva0110
Penile0110
Fallopian0110
Peritoneal101100.0
Hepatic0110
Total18545864328.7

Colorectal, breast, and lung cancers were the most common malignancies tested. Overall, 28.7% of patients demonstrated no FancD2 foci formation in their tumor specimens. FA functional deficiency was observed throughout most histological types tested.

Dose Escalation and Toxicities

When appropriate, FATSI-negative patients consented to the therapeutic portion of the study. Sixty-one patients were enrolled through one of 14 dose levels in the two arms. Table 2 depicts the demographics and tumor types of these patients.

Table 2.

Patient characteristics*

CharacteristicNo. of patients
Arm 1 (veliparib)Arm 2 (veliparib +MMC)All
No. of enrolled patients322961
Men/women, No.15/1715/1430/31
Race/ethnicity, No.
 White292847
 African American314
 Hispanic/Latino000
 Other000
 Median age (range), y57 (27–79)59 (30–77)58 (27–79)
ECOG Performance Status, No.
 0101323
 1201535
 2213
Primary tumor type, No.
 Breast6612
 Colorectal11920
 Non–small cell lung336
 Small cell lung202
 Ovarian314
 Small bowel022
 Pancreatic022
 Cholangiocarcinoma112
 Mesothelioma112
 Thymic, bladder, ampullary, anal, testicular505
 Nasopharyngeal, prostate, melanoma, endometrial044
Previous chemotherapy
 None000
 1 line369
 2 lines181432
 ≥3 lines11920
CharacteristicNo. of patients
Arm 1 (veliparib)Arm 2 (veliparib +MMC)All
No. of enrolled patients322961
Men/women, No.15/1715/1430/31
Race/ethnicity, No.
 White292847
 African American314
 Hispanic/Latino000
 Other000
 Median age (range), y57 (27–79)59 (30–77)58 (27–79)
ECOG Performance Status, No.
 0101323
 1201535
 2213
Primary tumor type, No.
 Breast6612
 Colorectal11920
 Non–small cell lung336
 Small cell lung202
 Ovarian314
 Small bowel022
 Pancreatic022
 Cholangiocarcinoma112
 Mesothelioma112
 Thymic, bladder, ampullary, anal, testicular505
 Nasopharyngeal, prostate, melanoma, endometrial044
Previous chemotherapy
 None000
 1 line369
 2 lines181432
 ≥3 lines11920

* MMC = mitomycin C.

Table 2.

Patient characteristics*

CharacteristicNo. of patients
Arm 1 (veliparib)Arm 2 (veliparib +MMC)All
No. of enrolled patients322961
Men/women, No.15/1715/1430/31
Race/ethnicity, No.
 White292847
 African American314
 Hispanic/Latino000
 Other000
 Median age (range), y57 (27–79)59 (30–77)58 (27–79)
ECOG Performance Status, No.
 0101323
 1201535
 2213
Primary tumor type, No.
 Breast6612
 Colorectal11920
 Non–small cell lung336
 Small cell lung202
 Ovarian314
 Small bowel022
 Pancreatic022
 Cholangiocarcinoma112
 Mesothelioma112
 Thymic, bladder, ampullary, anal, testicular505
 Nasopharyngeal, prostate, melanoma, endometrial044
Previous chemotherapy
 None000
 1 line369
 2 lines181432
 ≥3 lines11920
CharacteristicNo. of patients
Arm 1 (veliparib)Arm 2 (veliparib +MMC)All
No. of enrolled patients322961
Men/women, No.15/1715/1430/31
Race/ethnicity, No.
 White292847
 African American314
 Hispanic/Latino000
 Other000
 Median age (range), y57 (27–79)59 (30–77)58 (27–79)
ECOG Performance Status, No.
 0101323
 1201535
 2213
Primary tumor type, No.
 Breast6612
 Colorectal11920
 Non–small cell lung336
 Small cell lung202
 Ovarian314
 Small bowel022
 Pancreatic022
 Cholangiocarcinoma112
 Mesothelioma112
 Thymic, bladder, ampullary, anal, testicular505
 Nasopharyngeal, prostate, melanoma, endometrial044
Previous chemotherapy
 None000
 1 line369
 2 lines181432
 ≥3 lines11920

* MMC = mitomycin C.

Dose escalation, numbers of patients with dose reductions, DLT, and the MTD for each arm of the trial are depicted in Table 3 . Overall toxicities are depicted on Table 4 . For veliparib monotherapy, two of three patients developed DLT (grade 3 fatigue) at 400mg BID during their first cycle. The first subject at this level tolerated well one cycle but discontinued trial participation for surgical resection of his mesothelioma. However, a subject age 77 years with metastatic breast cancer developed grade 2 nausea/vomiting and diarrhea 24 hours after veliparib initiation, which improved by the following day with drug interruption. Reintroduction of veliparib resulted in reappearance of symptoms, development of grade 3 fatigue and consent withdrawal (received 8 doses). The other subject with DLT (a woman age 63 years with colon cancer) developed lightheadedness and worsening fatigue over the first cycle, which she completed, spending more than 50% of her day resting or in bed. Interruption of veliparib for two weeks resulted in improvement of fatigue to grade 2. Treatment was discontinued because of clinical disease progression.

Table 3.

Dose escalation and toxicities of veliparib alone and with MMC

Dose levelVeliparib mg MMC mg/m 2No. patients treatedNo. cyclesPts with DLT/ evaluablePts with dose delays/ reductions
Arm 1
 150 BID-3640/30
 280 BID-350/30
 3100 BID-3170/30
 4150AM/100PM-360/30
 5150 BID-380/30
 6200AM/150PM-4110/30
 7200 BID-370/30
 8300 BID-7150/60
 9400 BID-342 /30
Total--32137--
Arm 2*
 1A50 BID x 7d10 q 28d4110/30
 2A50 BID x 14d10 q 28d7230/6†0
 3A50 BID x 21d10 q 28d3110/30
 4A100 BID x21d10 q 28d4450/31/1
 5A200 BID x 21d10 q 28d11221/60/1
Total--29112--
Dose levelVeliparib mg MMC mg/m 2No. patients treatedNo. cyclesPts with DLT/ evaluablePts with dose delays/ reductions
Arm 1
 150 BID-3640/30
 280 BID-350/30
 3100 BID-3170/30
 4150AM/100PM-360/30
 5150 BID-380/30
 6200AM/150PM-4110/30
 7200 BID-370/30
 8300 BID-7150/60
 9400 BID-342 /30
Total--32137--
Arm 2*
 1A50 BID x 7d10 q 28d4110/30
 2A50 BID x 14d10 q 28d7230/6†0
 3A50 BID x 21d10 q 28d3110/30
 4A100 BID x21d10 q 28d4450/31/1
 5A200 BID x 21d10 q 28d11221/60/1
Total--29112--

* Two evaluable cycles required for dose-limiting toxicity evaluation. BID = twice daily; DLT = dose-limiting toxicity; MMC = mitomycin C; Pts = patients.

† Because of a G3 dyspnea episode, which was unclear if related to MMC or to tumor progression after a third cycle, the dose level was expanded to six patients.

Table 3.

Dose escalation and toxicities of veliparib alone and with MMC

Dose levelVeliparib mg MMC mg/m 2No. patients treatedNo. cyclesPts with DLT/ evaluablePts with dose delays/ reductions
Arm 1
 150 BID-3640/30
 280 BID-350/30
 3100 BID-3170/30
 4150AM/100PM-360/30
 5150 BID-380/30
 6200AM/150PM-4110/30
 7200 BID-370/30
 8300 BID-7150/60
 9400 BID-342 /30
Total--32137--
Arm 2*
 1A50 BID x 7d10 q 28d4110/30
 2A50 BID x 14d10 q 28d7230/6†0
 3A50 BID x 21d10 q 28d3110/30
 4A100 BID x21d10 q 28d4450/31/1
 5A200 BID x 21d10 q 28d11221/60/1
Total--29112--
Dose levelVeliparib mg MMC mg/m 2No. patients treatedNo. cyclesPts with DLT/ evaluablePts with dose delays/ reductions
Arm 1
 150 BID-3640/30
 280 BID-350/30
 3100 BID-3170/30
 4150AM/100PM-360/30
 5150 BID-380/30
 6200AM/150PM-4110/30
 7200 BID-370/30
 8300 BID-7150/60
 9400 BID-342 /30
Total--32137--
Arm 2*
 1A50 BID x 7d10 q 28d4110/30
 2A50 BID x 14d10 q 28d7230/6†0
 3A50 BID x 21d10 q 28d3110/30
 4A100 BID x21d10 q 28d4450/31/1
 5A200 BID x 21d10 q 28d11221/60/1
Total--29112--

* Two evaluable cycles required for dose-limiting toxicity evaluation. BID = twice daily; DLT = dose-limiting toxicity; MMC = mitomycin C; Pts = patients.

† Because of a G3 dyspnea episode, which was unclear if related to MMC or to tumor progression after a third cycle, the dose level was expanded to six patients.

Table 4.

Hematologic toxicities of veliparib alone and in combination with mitomycin C in FA pathway deficient patients

Toxicity*No. of patients’ cycles with toxicity by toxicity grade
Veliparip, all dosesVeliparib, MTDVeliparib-MMC, all dosesVeliparib-MMC, MTD
234234234234
Hematologic
 Neutropenia100000410000
 Anemia11215001640230
 Thrombocytopenia10010097†1530
 Lymphocytopenia133011016110870
 Febrile neutropenia000000000000
Nonhematologic
 Nausea610300510400
 Vomiting920300210110
 Anorexia210300700500
 Diarrhea720410310000
 Dysgeusia000000800800
 Headaches010000110010
 Transaminitis200000120100
 Alkaline phosphatase210000211210
 Fatigue/asthenia37022023901240
 Arthralgia/myalgia120100200200
 Peripheral neuropathy1000200700000
 Hyperglycemia310000301201
 Peripheral edema000000300300
Toxicity*No. of patients’ cycles with toxicity by toxicity grade
Veliparip, all dosesVeliparib, MTDVeliparib-MMC, all dosesVeliparib-MMC, MTD
234234234234
Hematologic
 Neutropenia100000410000
 Anemia11215001640230
 Thrombocytopenia10010097†1530
 Lymphocytopenia133011016110870
 Febrile neutropenia000000000000
Nonhematologic
 Nausea610300510400
 Vomiting920300210110
 Anorexia210300700500
 Diarrhea720410310000
 Dysgeusia000000800800
 Headaches010000110010
 Transaminitis200000120100
 Alkaline phosphatase210000211210
 Fatigue/asthenia37022023901240
 Arthralgia/myalgia120100200200
 Peripheral neuropathy1000200700000
 Hyperglycemia310000301201
 Peripheral edema000000300300

* Toxicities are reported as per National Cancer Institute common toxicity criteria 4. Two hundred forty-nine cycles were administered: 137 for veliparib, 15 for veliparib at maximum tolerated dose (MTD), 112 for mitomycin C (MMC)/veliparib, 22 for MMC/veliparib at MTD. FA = Fanconi Anemia; MMC = mitomycin C; MTD = maximum tolerated dose.

† One episode of thrombotic microangiopathy.

Table 4.

Hematologic toxicities of veliparib alone and in combination with mitomycin C in FA pathway deficient patients

Toxicity*No. of patients’ cycles with toxicity by toxicity grade
Veliparip, all dosesVeliparib, MTDVeliparib-MMC, all dosesVeliparib-MMC, MTD
234234234234
Hematologic
 Neutropenia100000410000
 Anemia11215001640230
 Thrombocytopenia10010097†1530
 Lymphocytopenia133011016110870
 Febrile neutropenia000000000000
Nonhematologic
 Nausea610300510400
 Vomiting920300210110
 Anorexia210300700500
 Diarrhea720410310000
 Dysgeusia000000800800
 Headaches010000110010
 Transaminitis200000120100
 Alkaline phosphatase210000211210
 Fatigue/asthenia37022023901240
 Arthralgia/myalgia120100200200
 Peripheral neuropathy1000200700000
 Hyperglycemia310000301201
 Peripheral edema000000300300
Toxicity*No. of patients’ cycles with toxicity by toxicity grade
Veliparip, all dosesVeliparib, MTDVeliparib-MMC, all dosesVeliparib-MMC, MTD
234234234234
Hematologic
 Neutropenia100000410000
 Anemia11215001640230
 Thrombocytopenia10010097†1530
 Lymphocytopenia133011016110870
 Febrile neutropenia000000000000
Nonhematologic
 Nausea610300510400
 Vomiting920300210110
 Anorexia210300700500
 Diarrhea720410310000
 Dysgeusia000000800800
 Headaches010000110010
 Transaminitis200000120100
 Alkaline phosphatase210000211210
 Fatigue/asthenia37022023901240
 Arthralgia/myalgia120100200200
 Peripheral neuropathy1000200700000
 Hyperglycemia310000301201
 Peripheral edema000000300300

* Toxicities are reported as per National Cancer Institute common toxicity criteria 4. Two hundred forty-nine cycles were administered: 137 for veliparib, 15 for veliparib at maximum tolerated dose (MTD), 112 for mitomycin C (MMC)/veliparib, 22 for MMC/veliparib at MTD. FA = Fanconi Anemia; MMC = mitomycin C; MTD = maximum tolerated dose.

† One episode of thrombotic microangiopathy.

The 300mg BID dose was expanded to six evaluable patients. At this dose, toxicities included one patient with grade 3 fatigue lasting fewer than seven days, a grade 3 pneumonia during cycle 2, and a grade 3 diarrhea during cycle 5. The 300mg BID veliparib dose was therefore declared the MTD for veliparib as monotherapy for patients with FA-deficient tumors.

For the combination arm, one patient receiving 14 days of veliparib dosing (2A) developed grade 3 fatigue, dyspnea, hypoxia, and a pleural effusion after completing two cycles. Because of the possibility of MMC-induced hypoxia, the dose level was expanded, with no additional moderate or severe toxicities. Two cycles were required subsequently for the combination arm as a safety precaution for evaluation of DLT. For patients receiving 21 days of veliparib (3A), no moderate/severe toxicities occurred during the first two cycles. However, one patient developed a thrombotic thrombocytopenic purpura-like syndrome after cycle 3. Veliparib dose in this schedule was subsequently escalated, up to 200mg BID. At this last dose level, the enrollment of 11 patients was necessary to assure that at least six patients completed two cycles. Although only one DLT (grade 3 fatigue) occurred, the inability to deliver MMC consistently on schedule after cycle 3 made additional dose escalation impractical. Thus MMC 10mg/m 2 followed by 200mg BID veliparib is recommended for initial dosing in subsequent trials.

Antitumor Activity

Supplementary Table 4 (available online) depicts tumor assessments results. Six antitumor responses were confirmed (5-RECIST/1-PET criteria) ( 37 , 38 ). Three patients withdrew consent during the first cycle without progression. Two withdrew because of ill effects, and the third to undergo an extrapleural pneumonectomy. Histologic sections demonstrated less than 10% residual viable tumor cells in the resected pleura. Two patients in the combination arm withdrew for side effects without progression after one cycle.

A patient with metastatic ovarian cancer who had pathological documentation of PET avid metastasis to liver after treatment with cisplatin/gemcitabine had resolution of PET uptake after two cycles of therapy at dose level 5A. Previous treatment included paclitaxel/carboplatin for ovarian cancer and adjuvant doxorubicin for a previous bilateral breast cancer. Partial responses occurred in three patients with breast cancer, two of whom are still receiving therapy (dose level 1, 60 cycles; and dose level 4A, 36 cycles), in a non–small cell lung cancer patient (1A), and in a patient with endometrial carcinoma metastatic to peritoneum (5A). Stable disease was the best response in 18 patients (median = 6 months, range = 3–15 months).

Correlative Studies

PBMC BRCA analysis (Myriad) among 51 patients receiving veliparib or veliparib/MMC showed that five patients (2 breast, 1 ampullary, 2 ovarian) carried BRCA-deleterious mutations ( Supplementary Table 5 , available online). Of note is the uncovering of four cases of breast cancer in the family of an ampullary carcinoma patient with a previously unsuspected BRCA2 deleterious mutation and the discovery of this mutation in two of his three daughters ( Figure 3 ).

Patient with ampullary carcinoma and BRCA2 mutation. Four cases of breast cancer in his family were identified. The testing of his three daughters identified the deleterious mutation in the germline of two of them (subjects 29 and 35).
Figure 3.

Patient with ampullary carcinoma and BRCA2 mutation. Four cases of breast cancer in his family were identified. The testing of his three daughters identified the deleterious mutation in the germline of two of them (subjects 29 and 35).

Seventeen patients receiving veliparib had PAR assessment on PBMC ( Supplementary Figure 1 , available online). Thirteen of 15 patients with evaluable data had reductions during cycle 1. PAR level was lower at cycle 2 predose timepoint than that of cycle 1 predose in six of seven patients on veliparib alone, suggesting long-term modulation of PARP activity. Following MMC, four of five patients had higher PAR levels at cycle 2 predose than that of C1, suggesting mitomycin-C activated PARP. However, PAR levels went down in all patients in subsequent samples.

Forty-seven patients were evaluated for gamma-H2AX on PBMC ( Supplementary Tables 2 and 3 , available online). Limited data points from informative patients of gama-H2AX cytospin made it difficult to draw any conclusion. Significant induction (>2%) only occurred in two patients in arm 1 and five from arm 2.

We sequenced 49 random patient tumor DNA samples, FATSI-negative per screening, with the FA sequencing panel. Thirty-four unique alterations were identified in 29 of the 49 patient specimens ( Supplementary Table 6 , available online). Seventeen patients who received veliparib were among those sequenced ( Supplementary Table 7 , available online). Tumor DNA from the two patients with germline BRCA mutations analyzed showed the same germline mutations at a high variant allele frequency (VAF). Loss of heterozigosity (LOH) was confirmed in the tumor. A RAD51c mutation (c.223_224insA p.Y75*) with high VAF was detected in the tumor from a breast cancer patient with a PR who was on maintenance veliparib for two years after initial induction with veliparib/MMC. The same RAD51c mutation was demonstrated in her germline tissue (Invitae Panel, San Francisco, CA), as well as family history consistent for a cancer predisposition syndrome ( Figure 4 ).

Family tree of patient with breast cancer and a Rad51c mutation in tumor and germline. Three cases of breast cancer, one of ovarian cancer, and one of leukemia in the family were detected.
Figure 4.

Family tree of patient with breast cancer and a Rad51c mutation in tumor and germline. Three cases of breast cancer, one of ovarian cancer, and one of leukemia in the family were detected.

One additional patient with metastatic lung adenocarcinoma and tracheal infiltration had a truncation mutation in the ataxia telangiectasia mutated gene (ATM c.6976-1 G>T), which was not present in his germline, in addition to an ERCC4 missense mutation (P379S) in both germline and tumor with LOH. An episode of massive hemoptysis occurred during his first cycle of veliparib monotherapy, requiring intubation. Continuation of life support was declined by the family.

Tumors and adjacent tissue from 10 patients FATSI-positive per screening were analyzed as controls with the FA sequencing panel. A deleterious mutation (ERCC4), along with a germline potentially damaging mutation in FAN1, was found in only one patient. One other patient had a somatic missense mutation in ATM ( Supplementary Table 6 , available online).

Discussion

The FATSI screening results confirm that a substantial number of patients throughout a variety of primary organ sites have FA functional deficiency in their tumors. Colorectal, breast, and lung cancers had the most representation. The 29% rate is consistent with our previously reported data in tumors obtained from the Cooperative Human Tissue Network ( 32 ). Although the Ki67 (a commonly used tumor proliferation index marker) expression requirement for the test (>10%) to be evaluable helps to eliminate false negatives because of necrosis, fibrosis, etc., low-proliferating tumors may be affected in a similar way, thus they are not suitable for this test.

The targeted FA sequencing panel was able to show FA-associated repair alterations at the genomic level in a substantial fraction of FATSI-negative patients, as compared with a control group of FATSI positives. Other mechanisms for foci formation loss not examined could include large deletions not readily captured by the panel, epigenetic silencing of the FA genes, or mutations in genes not included. To test this notion, an expansion cohort of FATSI-negative cancer patients is being accrued who will provide fresh tumor biopsies prior to treatment. Our prediction is that by adding RNAseq, we will be able to identify the cause of the functional defect detected by FATSI in most patients with no mutations on the repair sequencing panel. Therefore, it is our contention that the test could not only provide an inexpensive and practical tool for population screening for tumor-associated DNA-repair dysfunction, which then could be targeted for sequencing, but could also provide the phenotypic dysfunction readout of a yet-uncharacterized repair gene alteration identified by genetic screening.

Another dimension added by FATSI screening was that it led to identification of unsuspected germline mutations, which impacted screening and prevention measures in the families of some of our patients. These included an uncommon Rad51c insertion mutation in a breast cancer patient predicted to be pathogenic ( 39–40 ). Both ovarian and breast cancer were present in her genealogical tree. The same mutation was identified in both germline and tumor material from a sister recently diagnosed with ovarian cancer.

Our study also showed that it is safe to administer veliparib at doses of up to 300mg BID to FA-deficient patients and that veliparib can be safely combined with the DNA-breaking agent mitomycin-C. However, veliparib as monotherapy did not produce a substantial number of tumor regressions. Potential reasons would include: 1) veliparib spectrum of doses below MTD utilized (it should also be noted that doses of up to 400mg BID have been tolerated in a different population of patients); 2) veliparib relatively low PARP trapping activity (a newly described mechanism of action for PARPi) ( 41 , 42 ); 3) restoration of FA functionality as a result of systemic treatment in the interval between the material in archives and enrollment in the trial ( 43 , 48 ); or 4) the presence of an additional antiapoptotic stimulus to which the repair deficient cell has become addicted (nononcogenic addiction/induced-essentiality) ( 49 , 50 ). The latter mechanism is expected to operate more commonly in non-BRCA-mutated, repair-deficient tumors, as BRCA mutation is in itself a potent anti-apoptotic stimulus.

A few genomic signatures have come forward as suggestive of “BRCAness” in tumors, and evaluations of their potential for enriching patient populations most sensitive to PARPi are actively being pursued ( 51–54 ). Given that FATSI is a functional, rather than a genomic, analysis, it would be of great interest to explore correlations or overlap between these signatures and FATSI. Properly powered prospective therapeutic trials to evaluate concurrently FATSI and other potential biomarkers as predictors of PARPi tumor sensitivity at proper doses across diverse malignancies would help to clarify their comparative clinical value.

Limitations of our study include the lack of fresh biopsies to cross-validate and assess potential changes in the pathway activation induced by chemotherapy given in the interval between the available archival material and the initiation of the pharmacological intervention and the inability to assess well the functionality in low-proliferating tumors.

Future plans include an ongoing expansion cohort of the combination of veliparib/MMC at recommended doses in FATSI-negative colorectal cancer patients, in which fresh biopsies are being obtained for patient-derived tumor xenografts creation, in order to address and overcome resistance. A follow-up phase II clinical trial proposal has been endorsed by the NCI to test the ability of a potent PARP trapping agent ( 55 , 56 ) to achieve synthetic lethality in nonbreast/nonovarian FA-deficient patients. Patients are selected by either FATSI (1 cohort) or genomically defined cohorts harboring mutations pertinent to homologous recombination ( 58 ), De novo and acquired resistance will be evaluated. Fresh biopsies, cross validation between functional and genomic abnormalities, and whole-exome tumor sequencing will be performed in all patients.

Funding

This work was supported by National Institutes of Health (NIH) grants R01-CA152101, N01-CM-2011-00070 (HHSN261201100070C) to MAV, and OSUCCC P30 CA016058-38.

Notes

The study funders had no role in design of the study; the collection, analysis, or interpretation of the data; the writing of the manuscript; or the decision to submit the manuscript for publication.

The authors revealed no conflict of interests related to the performance of the work reported or the writing of this manuscript.

Special thanks at the OSU Molecular Core Facility to Kristin Kovach (FATSI) and Nehad Mohamed (KRAS and BRAF); at the OSUCCC Clinical Trial Office to Kirsten Keinsenmayer (outside tissue coordination and reporting) and Andrea Lively (regulatory); at Tissue Archives to Mariya Kravets; at Cancer Genetics to Robert Pilarski; and Yiping Zhang (NCI), Li Gao, and Britanny Aguila (MAV’s lab, preparation, and performance of the correlative assays) for their diligent work.

References

1.

Bryant
HE
Schultz
N
Thomas
HD
et al.
Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase
.
Nature
.
2005
;
434
(
7035
):
913
917
.

2.

Farmer
H
McCabe
N
Lord
CJ
et al.
Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy
.
Nature
.
2005
;
434
(
7035
):
917
921
.

3.

Tutt
A
Robson
M
Garber
J
et al.
Phase II trial of the oral PARP inhibitor olaparib in BRCA-deficient advanced breast cancer
.
J Clin Oncol
.
2009
;
27
(
Suppl
;
Abstr CRA501
):
18s
.

4.

Fong
P
Boss
D
Yap
T
et al.
Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers
.
N Engl J Med
.
2009
;
361
(
2
):
123
134
.

5.

Audeh
W
Carmichael
J
Penson
RT
et al.
Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer: a proof-of-concept trial
.
Lancet
.
2010
;
376
(
9737
):
245
251
.

6.

Bagby
GC
Jr .
Genetic basis of Fanconi anemia
.
Curr Opin Hematol
.
2003
;
10
(
1
):
68
76
.

7.

D’Andrea
AD
Grompe
M
.
The Fanconi anaemia/BRCA pathway
.
Nat Rev Cancer
.
2003
;
3
:
23
34
.

8.

Reid
S
Schindler
D
Hanenberg
H
et al.
Biallelic mutations in PALB2 cause Fanconi anemia subtype FA-N and predispose to childhood cancer
.
Nat Genet
.
2007
;
39
(
2
):
162
164
.

9.

Xia
B
Dorsman
JC
Ameziane
N
et al.
Fanconi anemia is associated with a defect in the BRCA2 partner PALB2
.
Nat Genet
.
2007
;
39
(
2
):
159
161
.

10.

Smogorzewska
A
Matsuoka
S
Vinciguerra
P
et al.
Identification of the FANCI protein, a monoubiquitinated FANCD2 paralog required for DNA repair
.
Cell
.
2007
;
129
(
2
):
289
301
.

11.

Kim
Y
Lach
FP
Desetty
R
et al.
SLX4, a coordinator of structure-specific endonucleases, is mutated in a new Fanconi anemia subtype
.
Nat Genet
.
2011
;
43
:
138
141
.

12.

Vaz
F
Hanenberg
H
Schuster
B
et al.
Mutation of the RAD51C gene in a Fanconi anemia-like disorder
.
Nat Genet
.
2010
;
42
:
406
409
.

13.

Machida
YJ
Machida
Y
Chen
Y
et al.
UBE2T is the E2 in the Fanconi anemia pathway and undergoes negative autoregulation
.
Mol Cell
.
2006
;
23
(
4
):
589
596
.

14.

Meetei
AR
Yan
Z
Wang
W
.
FANCL replaces BRCA1 as the likely ubiquitin ligase responsible for FANCD2 monoubiquitination
.
Cell Cycle
.
2004
;
3
(
2
):
179
181
.

15.

Tischkowitz
M
Xia
B
Sabbaghian
N
et al.
Analysis of PALB2/FANCN-associated breast cancer families
.
Proc Natl Acad Sci U S A
.
2007
;
104
(
16
):
6788
6793
.

16.

Garcia-Higuera
I
Taniguchi
T
Ganesan
S
et al.
Interaction of the Fanconi anemia proteins and BRCA1 in a common pathway
.
Mol Cell
.
2000
;
7
(
2
):
249
262
.

17.

Stoepker
C
Hain
K
Schuster
B
et al.
SLX4, a coordinator of structure-specific endonucleases, is mutated in a new Fanconi anemia subtype
.
Nat Genet
.
2011
;
43
:
138
141
.

18.

Bogliolo
M
Schuster
B
Stoepker
C
et al.
Mutations in ERCC4, encoding the DNA-repair endonuclease XPF, cause Fanconi anemia
.
Am J Hum Genet
.
2013
;
92
(
5
):
800
806
.

19.

Sawyer
SL
Tian
L
Kähkönen
M
et al.
Biallelic mutations in BRCA1 cause a new Fanconi Anemia subtype
.
Cancer Discov
.
2015
;
5
(
2
):
135
142
.

20.

Kwee
ML
Poll
EH
van de Kamp
JJ
de Koning
H
Eriksson
AW
Joenje
H
.
Unusual response to bifunctional alkylating agents in a case of Fanconi anaemia
.
Hum Genet
.
1983
;
64
(
4
):
384
387
.

21.

Auerbach
AD
Rogatko
A
Schroeder-Kurth
TM
.
International Fanconi Anemia Registry: relation of clinical symptoms to diepoxybutane sensitivity
.
Blood
.
1989
;
73
(
2
):
391
396
.

22.

Alter
BP
.
Cancer in Fanconi anemia, 1927–2001
.
Blood
.
2003
;
101
(
5
):
2072
.

23.

Thompson
LH
Hinz
JM
.
Cellular and molecular consequences of defective fanconi anemia proteins in replication-coupled DNA repair: mechanistic insights
.
Mutational Res
.
2009
;
668
:
54
72
.

24.

Lyakhovich
A
Surralles
J
.
Disruption of the Fanconi anemia/BRCA pathway in sporadic cancer
.
Cancer Lett
.
2006
;
232
(
1
):
99
106
.

25.

Neveling
K
Kalb
R
Florl
AR
et al.
Disruption of the FA/BRCA pathway in bladder cancer
.
Cytogenet Genome Res
.
2007
;
118
(
2
4
):
166
176
.

26.

Taniguchi
T
Tischkowitz
M
Ameziane
N
et al.
Disruption of the Fanconi anemia-BRCA pathway in cisplatin-sensitive ovarian tumors
.
Nat Med
.
2003
;
9
(
5
):
568
574
.

27.

Zhang
J
Wang
X
Lin
CJ
Couch
FJ
Fei
P
.
Altered expression of FANCL confers mitomycin C sensitivity in Calu-6 lung cancer cells
.
Cancer Biol Ther
.
2006
;
5
(
12
):
1632
1636
.

28.

van der Heijden
MS
Brody
JR
Dezentje
DA
et al.
In vivo therapeutic responses contingent on Fanconi anemia/BRCA2 status of the tumor
.
Clin Cancer Res
.
2005
;
11
(
20
):
7508
7515
.

29.

Moiseyenko
VM
Chubenko
VA
Moiseyenko
FV
et al.
Evidence for clinical efficacy of mitomycin C in heavily pretreated ovarian cancer patients carrying germ-line BRCA1 mutation
.
Med Oncol
.
2014
;
31
(
10
):
199
.

30.

Sonnenblick
A
Kadouri
L
Appelbaum
L
et al.
Complete remission, in BRCA2 mutation carrier with metastatic pancreatic adenocarcinoma, treated with cisplatin based therapy
.
Cancer Biol Ther
.
2011
;
12
(
3
):
165
168
.

31.

Tutt
A
Ellis
P
Kilburn
L
et al.
The TNT trial: A randomized phase III trial of carboplatin (C) compared with docetaxel (D) for patients with metastatic or recurrent locally advanced triple negative or BRCA1/2 breast cancer (CRUK/07/012)
.
San Antonio Breast Cancer Symposium
2014
;
S3
S01
.

32.

Duan
W
Gao
L
Zhao
W
et al.
Assesment of FANCD2 nuclear foci formation in paraffin embedded tumors: a potential patient enrichment strategy for treatment with DNA interstrand crosslink agents
.
Transl Res
.
2013
;
161
(
3
):
156
164
.

33.

Duan
W
Gao
L
Aguila
B
Kalvala
A
Otterson
GA
Villalona-Calero
MA
.
Fanconi anemia repair pathway dysfunction, a potential therapeutic target in lung cancer
.
Front Oncol
.
2014
;19;
4
:
368
.

34.

Kummar
S
Kinders
R
Gutierrez
ME
et al.
Phase 0 clinical trial of the poly (ADP-ribose) polymerase inhibitor ABT-888 in patients with advanced malignancies
.
J Clin Oncol
.
2009
;
27
(
16
):
2705
2711
.

35.

Redon
C
Nakamura
A
Zhang
Y
et al.
Histone γH2AX and Poly(ADP-Ribose) as Clinical Pharmacodynamic Biomarkers
.
Clin Cancer Res
.
2010
;
16
:
4532
.

36.

Araujo
L
Timmers
CD
Hlavin-Bell
E
et al.
Genomic characterization of non-small cell lung cancer in African Americans by targeted massively parallel sequencing
.
J Clin Oncol
.
2015
;
33
(
17
):
1966
1973
.

37.

Eisenhauer
E
Therasse
P
Bogaerts
J
et al.
New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1)
.
Eur J Cancer
.
2009
;
45
:
228
247
.

38.

Shankar
LK
Hoffman
JM
Bacharach
S
et al.
Consensus recommendations for the use of 18F-FDG PET as an indicator of therapeutic response in patients in National Cancer Institute Trials
.
J Nucl Med
.
2006
;
47
:
1059
1066
.

39.

Meindl
A
Hellebrand
H
Wiek
C
et al.
Germline mutations in breast and ovarian cancer pedigrees establish RAD51C as a human cancer susceptibility gene
.
Nat Genet
.
2010
;
42
(
5
):
410
414
.

40.

González-Pérez
A
López-Bigas
N
.
Improving the Assessment of the Outcome of Nonsynonymous SNVs with a Consensus Deleteriousness Score, Condel
.
Am J Hum Genet
.
2011
;
88
:
440
449
.

41.

Murai
J
Huang
SY
Das
BB
et al.
Trapping of PARP1 and PARP2 by Clinical PARP Inhibitors
.
Cancer Res
.
2012
;
72
(
21
):
5588
5599
.

42.

Murai
J
Huang
SY
Renaud
A
et al.
Stereospecific PARP trapping by BMN 673 and comparison with olaparib and rucaparib
.
Mol Cancer Ther
.
2014
;
13
(
2
):
433
443
.

43.

Lord
CJ
Ashworth
A
.
Mechanisms of resistance to therapies targeting BRCA-mutant cancers
.
Nat Med
.
2013
;
19
(
11
):
1381
1388
.

44.

Swisher
EM
Sakai
W
Karlan
BY
Wurz
K
Urban
N
Taniguchi
T
.
Secondary BRCA1 mutations in BRCA1-mutated ovarian carcinomas with platinum resistance
.
Cancer Res
.
2008
;
68
(
8
):
2581
2586
.

45.

Norquist
B
Wurz
KA
Pennil
CC
et al.
Secondary somatic mutations restoring BRCA1/2 predict chemotherapy resistance in hereditary ovarian carcinomas
.
J Clin Oncol
.
2011
;
29
(
22
):
3008
3015
.

46.

Barber
LJ
Sandhu
S
Chen
L
et al.
Secondary mutations in BRCA2 associated with clinical resistance to a PARP inhibitor
.
J Pathol
.
2013
;
229
(
3
):
422
429
.

47.

Cao
L
Xu
X
Bunting
SF
et al.
A selective requirement for 53BP1 in the biological response to genomic instability induced by Brca1 deficiency
.
Mol Cell
.
2009
;
35
(
4
):
534
541
.

48.

Bouwman
P
Aly
A
Escandell
JM
et al.
53BP1 loss rescues BRCA1 deficiency and is associated with triple-negative and BRCA-mutated breast cancers
.
Nat Struct Mol Biol
.
2010
;
17
(
6
):
688
695
.

49.

Luo
J
Somilini
NL
Elledge
SJ
.
Principles of cancer therapy: Oncogene and non-oncogene addiction
.
Cell
.
2009
;
136
:
823
837
.

50.

Tischler
J
Lehner
B
Frazer
AG
.
Evolutionary plasticity of genetic interaction networks
.
Nat Gen
.
2008
;
40
:
390
391
.

51.

Daemen
A
Wolf
DM
Korkola
JE
et al.
Cross-platform pathway-based analysis identifies markers of response to the PARP inhibitor olaparib
.
Breast Cancer Res Treat
.
2012
;
135
(
2
):
505
517
.

52.

Ihnen
M
zu Eulenburg
C
Kolarova
T
et al.
Therapeutic potential of the poly(ADP-ribose) polymerase inhibitor rucaparib for the treatment of sporadic human ovarian cancer
.
Mol Cancer Ther
.
2013
;
12
(
6
):
1002
1015
.

53.

Watkins
JA
Irshad
S
Grigoriadis
A
Tutt
AN
.
Genomic scars as biomarkers of homologous recombination deficiency and drug response in breast and ovarian cancers
.
Breast Cancer Res
.
2014
;3;
16
(
3
):
211
.

54.

Wolf
DM
Yau
C
Sanil
A
et al.
Evaluation of an in vitro derived signature of olaparib response (PARPi-7) as a predictive biomarker of response to veliparib/carboplatin plus standard neoadjuvant therapy in high risk breast cáncer: Results from the I-SPY 2 Trial
.
San Antonio Breast Cancer Symposium
.
2014
;P3-06-05.

55.

Shen
Y
Rehman
FL
Feng
Y
et al.
BMN 673, a novel and highly potent PARP1/2 inhibitor for the treatment of human cancers with DNA repair deficiency
.
Clin Cancer Res
.
2013
;
19
(
18
):
5003
5015
.

56.

First-in-human trial of novel oral PARP inhibitor BMN 673 in patients with solid tumors
.
J Clin Oncol Suppl
.
2013
;
31
:Abstr
2580
.

57.

McCabe
N
Turner
NC
Lord
CJ
et al.
Deficiency in the repair of DNA damage by homologous recombination and sensitivity to poly(ADP-ribose) polymerase inhibition
.
Cancer Res
.
2006
;
66
(
16
):
8109
8115
.

58.

Pennington
KP
Walsh
T
Harell
MI
et al.
Germline and somatic mutations in homologous recombination genes predict platinum response and survival in ovarian, fallopian tube, and peritoneal carcinomas
.
Clin Cancer Res
.
2014
;
20
(
3
):
764
775
.

Supplementary data