Skip to main content
Therapeutic Advances in Medical Oncology logoLink to Therapeutic Advances in Medical Oncology
. 2024 Mar 30;16:17588359241242406. doi: 10.1177/17588359241242406

Exceptional synergistic response of PARP inhibitor and immune checkpoint inhibitor in esophageal adenocarcinoma with a germline BRCA2 mutation: a case report

Himil Mahadevia 1, Ben Ponvilawan 2, Ammar Al-Obaidi 3, Jennifer Buckley 4, Janakiraman Subramanian 5, Dhruv Bansal 6,
PMCID: PMC10981852  PMID: 38559611

Abstract

Immune checkpoint inhibitors (ICIs) and poly (ADP-ribose) polymerase (PARP) inhibitors have shown efficacy in various tumors. A significant therapeutic challenge with either ICIs or PARP inhibitors as monotherapy is treatment failure from intrinsic primary resistance or the development of secondarily acquired resistance after a period of responsiveness. The combination of PARP inhibitors and ICIs could mitigate this by potentiating treatment response. We describe an 83-year-old male patient who initially presented with abdominal pain, and weight loss along with alternating constipation and diarrhea. Imaging and biopsy revealed metastatic esophageal adenocarcinoma. Genomic testing revealed germline BRCA2 mutation. The patient initially underwent a few cycles of chemoimmunotherapy. However, due to intolerance to chemotherapy, the patient’s case was discussed at a multidisciplinary molecular tumor board. He was switched to PARP inhibitor olaparib and ICI nivolumab. This combination led to a durable complete response. A combination of poly-ADP ribose polymerase inhibitor (PARPi) plus ICI may work in synergy through various mechanisms including enhanced neoantigen expression, release of immune-activating cytokines, and increased programmed death-ligand 1 expression. This may culminate in accentuated efficacy outcomes with a manageable safety profile. This exceptional response with ICI and PARPi in our case is consistent with the synergistic value of this combination, and prospective studies are warranted to definitively characterize clinical utility.

Keywords: BRCA, combination therapy, DNA repair, esophageal cancer, homologous recombination, immune checkpoint inhibitor, PARP

Introduction

Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of multiple cancers.13 Pembrolizumab and nivolumab have exhibited improved survival when combined with systemic chemotherapy in the first-line treatment for unresectable esophageal cancer.46 More recently, poly (ADP-ribose) polymerase (PARP) inhibitors have also demonstrated significant activity in several cancer types, such as breast, ovarian, pancreatic, and prostate cancer with homologous recombination deficiency (HRD), such as those with BRCA1 or BRCA2 mutations.710 Several ongoing clinical trials are investigating the benefit of PARP inhibitors in this clinical setting.11,12 Despite these successes, many patients do not show long-term responses to PARP inhibitor monotherapy or in combination with chemotherapy. Most patients who initially responded to single-agent PARP inhibitor or ICI therapy eventually develop resistance and experience clinical progression.13,14

Growing evidence shows that PARP inhibitors and ICIs could synergistically act against tumor evasion via multiple mechanisms. Persistent DNA damage induced by PARP inhibition leads to potentiation of the immune response through increased expression of tumor neoantigens, enhancement of programmed cell death-ligand 1 (PD-L1) expression, activation of immune-activating pathways such as cGAS–STING (cyclic GMP-AMP synthase – stimulator of interferon genes) pathway, and modulation of the tumor immune microenvironment toward more aggressive T helper (TH)1 state.1519 These mechanisms could enhance the immune system and accentuate the rate and durability of response from ICIs, making a combination of ICIs and PARP inhibitors an attractive option for tumors enriched in an immunosuppressive tumor microenvironment, including esophageal cancer. 20 Here, we describe an exceptional case of metastatic esophageal adenocarcinoma who achieved complete response with combination therapy of programmed cell death-1 (PD-1) inhibitor nivolumab and PARP inhibitor olaparib.

Case presentation

An 83-year-old male with a past medical history of type 2 diabetes, hypertension, diverticulitis, and basal cell carcinoma presented with 4 months of ongoing abdominal pain, 10 lb weight loss, intermittent constipation, and diarrhea. His baseline [Eastern Cooperative Oncology Group (ECOG)] performance status was 1. The initial workup was unremarkable, including a complete blood count and comprehensive metabolic profile. Contrast-enhanced computed tomography (CT) scan of the abdomen and pelvis demonstrated an abnormal mass-like soft tissue thickening and edema at the gastroesophageal (GE) junction, abnormal necrotic lymph nodes in the celiac axis, and hypoenhancing hepatic lesions. CT chest demonstrated multiple sub-centimeter pulmonary nodules. Magnetic resonance imaging of the abdomen with and without contrast corroborated these findings. A subsequent whole-body positron emission tomography (PET) scan revealed hypermetabolic activity associated with the GE junction mass, upper abdominal adenopathy, and liver lesions [Figure 1(a)].

Figure 1.

Figure 1.

PET scans at diagnosis and 4 months after PARP inhibitor and ICI therapy. (a) Axial fused images from PET/CT prior to treatment show hypermetabolic primary mass at GEJ (left), hypermetabolic metastatic upper abdominal lymphadenopathy, and right hepatic lesion (right). (b) Axial fused images from PET/CT after eight cycles of treatment with nivolumab and olaparib show complete resolution of the primary GEJ mass (left) as well as the metastatic upper abdominal lymphadenopathy and right hepatic lesion (right).

CT, computed tomography; GEJ, gastroesophageal junction; ICI, immune checkpoint inhibitor; PARP, poly (ADP-ribose) polymerase; PET, positron emission tomography.

Esophagogastroduodenoscopy (EGD) revealed an ulcerated mass extending from the lower third of the esophagus into the GE junction and posterior stomach body. The biopsy of the esophageal mass revealed adenocarcinoma with human epidermal growth factor receptor 2 (HER2) expression 2+ on immunohistochemistry (IHC) and equivocal by fluorescence in situ hybridization (FISH). Next-generation sequencing (NGS) of the tissue (via Tempus xT) revealed pathogenic BRCA2 mutations (germline c.3545_3546del p.F1182fs Frameshift – LOF and somatic c.2641G>T p.E881 – Stop gain – LOF), and somatic SMAD4, APC, and PHLPP1 mutations. The tumor was microsatellite stable, tumor mutational burden (TMB) was 4.2 mutations per megabase, tumor proportion score (TPS) was 5%, and combined positive score (CPS) was 5. Cell-free DNA (cfDNA) (via Tempus xF) analysis revealed similar findings – BRCA2 (c.3545_3546del p. F1182fs Frameshift – LOF and somatic c.2641G>T p.E881 – Stop gain – LOF) in addition to SMAD4 and APC mutations. Liver biopsy was then pursued, which revealed moderately differentiated adenocarcinoma, positive for CK7 and CDX2 and negative for CK20 and TTF-1, HER2 IHC expression 0. Hence, the patient was diagnosed with Siewert type II esophageal adenocarcinoma with biopsy-proven liver metastases.

The patient was initiated on palliative leucovorin, 5-fluorouracil, oxaliplatin (FOLFOX), and nivolumab. Despite dose reductions, he was poorly tolerant of chemotherapy and developed grade 3 fatigue, grade 3 anorexia, grade 2 diarrhea, and grade 1 nausea per CTCAE v5.0 criteria. He started having frequent falls and became wheelchair-bound. After six cycles of chemoimmunotherapy, a CT scan of the chest, abdomen, and pelvis revealed stable disease per RECIST 1.1 criteria.

Given the declining performance status and poor tolerance of chemotherapy, the case was presented at the multidisciplinary precision oncology conference at Saint Luke’s Cancer Institute, and the decision was made to initiate the combination of a PARP inhibitor and ICI. The patient was subsequently initiated on nivolumab 240 mg intravenously every 2 weeks in addition to olaparib orally 300 mg twice daily.

After two cycles, he developed grade 2 transaminitis (aspartate aminotransferase, 107; alanine transaminase, 73; and alkaline phosphatase, 101), and nivolumab was held for 2 weeks before being resumed after the resolution of transaminitis without any intervention. Two months after the initiation of treatment with the combination of PARP inhibitor and ICI, minimal residual disease (MRD) testing using a commercially available tumor-informed circulating tumor DNA (ctDNA) assay (Signatera MRD assay) was performed and it showed undetectable ctDNA levels, indicating a complete response. After eight cycles (4 months) of treatment with olaparib and nivolumab, a skull base to mid-thigh PET scan was performed which revealed a complete resolution of all previously noted hypermetabolic lesions with no evidence of new fluorodeoxyglucose avid lesions [Figure 1(b)]. After 5 months of treatment with this combination, nivolumab dosing was changed to 480 mg administered every 4 weeks, while olaparib was continued at the same dosage. An EGD with biopsy was performed after 10 months of treatment with the ICI and PARP inhibitor combination, with esophageal biopsies confirming a complete pathological response. Follow-up imaging and serial ctDNA MRD testing 18 months after diagnosis continued to indicate complete response. The patient experienced no additional side effects and returned to his baseline performance status. Thus, the patient has had a sustained complete response for around one and a half years and is still ongoing. The patient’s timeline from diagnosis until the last follow-up is delineated briefly in Figure 2.

Figure 2.

Figure 2.

Timeline of events.

The figure shows the timeline of events from diagnosis to last follow-up.

ctDNA, circulating tumor DNA; FOLFOX, 5-fluorouracil plus oxaliplatin; MRD, minimal residual disease; NGS, next-generation sequencing.

Discussion

DNA damage repair is a complex process involving several pathways and genes. 21 PARP inhibition interferes with the base excision repair (BER) pathway, which induces the formation of single-strand breaks (SSBs) in DNA. 22 Persistently unrepaired SSBs and the inability of autoPARylation (hindering the regular detachment of PARP enzymes from the DNA replication fork) further contribute to the formation of double-stranded breaks (DSBs) in DNA. In cancer cells with HRD, especially those with loss of function of BRCA1 or BRCA2, there is an inadequate repair of DSBs. This leads to the accumulation of DSBs, which culminates in cellular stress and ultimately results in cancer cell death, a phenomenon called ‘synthetic lethality’. 23

ICIs have proven to be an effective therapeutic approach in various cancers by blocking the interaction between immune checkpoint proteins and their receptors and allowing enhanced immune activation and, ultimately, a more powerful anti-tumor response. 24 Unfortunately, most patients have progressive disease or relapse with ICI monotherapy. 13 Multiple mechanisms have been proposed for the loss of response to ICI. First, insufficient neoantigens may lead to a weaker immune response as less repertoire can be primed to attack the tumor. 25 Second, excessive secretion of immunoregulatory cytokines and metabolic inhibitors, such as interleukin-19 (IL-19), transforming growth factor-β (TGF-β), and kynurenine, also inhibits immune cell activity. 25 Third, tumor microenvironments that favor tumor immunosuppression, including the inadequate infiltration of T effector cells, excessive regulatory T cells, and over-expression of inhibitory signaling pathways, could inhibit cytotoxic T cells from mounting immune responses against tumor cells. 25

PARPi could overcome this loss of immune effect from ICIs via several pathways. Tumors with HRD have been associated with higher mutational load and thus enhanced neoantigen accumulation, culminating in more antigenic peptide presentation, increased T-cell priming, and a stronger immune response against cancer. 11 PARP inhibitors create DSBs and higher expression of neoantigens, which may potentiate immune response. 26 Furthermore, PARP inhibitors have been shown to activate the cyclic GMP-AMP synthase (cGAS)-STING pathway.15,19 This pathway facilitates interferon type I response, which enhances antigen presentation by increasing the immune proteasomal activity in antigen-presenting cells. 27

Moreover, it instigates the release of chemokines, such as CXCL10 and CCL5, that enhance T-cell chemotaxis and increase peri-tumoral T-cell infiltration. 27 PARP inhibitors have also been shown to upregulate PD-L1 expression by inactivating glycogen synthase kinase 3β, a glycogen metabolism modulator, in a dose-dependent manner via the inhibition of proteasomal degradation.17,28 PARP inhibitors may also induce elevated PD-L1 expression by altering DNA damage response via the ATM-ATR-Chk1 pathway. 29 Though modest, PARP inhibitors may also switch the immune effect from chronic low-level inflammation to a more aggressive TH1 immune response. 16 In summary, with multiple potential mechanisms of immune enhancement, PARP inhibitors may accentuate response with ICIs and increase the durability of response.

Multiple clinical studies have established the benefit of ICIs in the first or later lines of treatment for esophageal cancer. Multiple phase III randomized-controlled trials (RCTs) utilizing ICIs showed statistically significant improvements in survival outcomes for the first-line treatment. CheckMate 648 compares nivolumab plus chemotherapy (CMT), nivolumab plus ipilimumab, and CMT alone in advanced esophageal squamous cell carcinoma (ESCC). ICI-containing regimens were shown to have superior overall survival (OS) and progression-free survival (PFS) regardless of PD-L1 expression. 30 CheckMate 649 compared nivolumab plus CMT versus CMT alone in advanced gastric cancer (GC), gastroesophageal junction (GEJ) cancer, and esophageal adenocarcinoma. The combination of nivolumab and CMT had significantly better OS and PFS than CMT alone, especially in those with PD-L1 CPS ⩾ 5. 4 KEYNOTE-590 is a study comparing pembrolizumab plus CMT versus placebo plus CMT in advanced ESCC and esophageal adenocarcinoma, which demonstrated significantly improved OS and PFS with pembrolizumab compared to chemotherapy in patients who had a PD-L1 CPS ⩾ 10, regardless of cell type. 5 KEYNOTE-181 is a phase III RCT comparing pembrolizumab versus CMT in advanced ESCC and esophageal adenocarcinoma. OS benefit was observed only in patients who had ESCC with PD-L1 CPS ⩾ 10. 31 Finally, ATTRACTION-3 is a phase III RCT comparing nivolumab versus CMT for individuals with advanced ESCC, which showed superior OS but not PFS for individuals treated with nivolumab. 32

By contrast, a limited number of studies have been conducted to determine the clinical utility of PARP inhibitors in GE cancer. A phase III RCT (GOLD trial) compared olaparib plus paclitaxel versus placebo plus paclitaxel in GC or GEJ as later-line therapy. 33 Unfortunately, the combination of olaparib and paclitaxel did not meet its primary endpoint of improving OS, including in patients with ATM mutation. 33 Despite the negative result, there is still value in determining the population that might benefit from PARP inhibitors in this setting, as there is emerging evidence that esophageal cancer could harbor genomic alterations involving the homologous recombination pathway.

A germline mutational analysis of 10,389 cases from The Cancer Genome Atlas (TCGA) PanCanAtlas cohort revealed that 1.09% of all cases with esophageal cancer carried germline BRCA2 mutation, slightly higher than the prevalence in all cancers combined (0.83%). In addition, of all cases from TCGA with a germline BRCA2 mutation, esophageal cancer constituted 2.33% of the cases [Figure 3(a) and (b)]. 34 This population of patients with germline BRCA mutations might benefit from PARP inhibitor therapy, and several ongoing trials using PARP inhibitors with or without CMT or ICIs for GE cancer are being tested to determine the efficacy of these treatment regimens (Table 1).

Figure 3.

Figure 3.

Prevalence of germline BRCA2 mutation (a) compared to the total number of cases in a particular type of cancer (b) compared to the total number of cases with germline BRCA2 mutations.

CM, cutaneous melanoma; EC, esophageal carcinoma; IBC, invasive breast carcinoma; OV, ovarian carcinoma; PAC, pancreatic adenocarcinoma; PRAD, prostate adenocarcinoma.

Table 1.

Ongoing studies with treatment using PARP inhibitors with or without ICI in gastroesophageal cancer.

PARPi ClinicalTrials.gov identifier Phase Population Estimated number of patients Treatment regimen Primary outcome Status
Olaparib NCT04592211 I/II Second-line recurrent/advanced GC/GEJ with HRR mutation and MSS 71 Olaparib + pembrolizumab + paclitaxel PFS, dose-limiting toxicity Not yet recruiting
NCT03008278 I/II Second-line recurrent/metastatic GC/GEJ 49 Olaparib + ramucirumab ORR, dose-limiting toxicity Active, not recruiting
Niraparib NCT03840967 II Second-line advanced/metastatic GC/GEJ/EAC with HRD or LOH-high 43 Niraparib ORR Active, not recruiting
Rucaparib NCT03995017 I/II Second-line advanced/metastatic GC/GEJ/EAC 34 Rucaparib + ramucirumab ± nivolumab ORR Active, not recruiting
Talazoparib NCT04511039 I Second-line advanced/metastatic GC/GEJ/EAC and CRC 21 Talazoparib + trifluridine/tipiracil Adverse events Recruiting

CRC, colorectal cancer; EAC, esophageal adenocarcinoma; GC, gastric cancer; GEJ, gastroesophageal cancer; HRD, homologous recombination deficiency; HRR, homologous recombination repair; ICI, immune checkpoint inhibitor; LOH, loss of heterozygosity; MSS, microsatellite stable; ORR, objective response rate; PARP, poly (ADP-ribose) polymerase; PFS, progression-free survival.

Based upon the possible synergistic effect of PARP inhibitors and ICIs, there has been some progress in translating this combination treatment for other types of cancer. A phase I/II MEDIOLA basket trial studied the effectiveness of olaparib and durvalumab combination in advanced small-cell lung cancer, breast cancer, ovarian cancer, and GC. For patients with relapsed GC, the overall response rate (ORR) and the 3-month disease control rate (DCR) were 10% and 26%, respectively. The median duration of response was 11.1 months. 35 Similar efficacy was also found in patients with small-cell lung cancer. 36 Of note, patients with platinum-sensitive relapsed ovarian cancer and germline BRCA1/2-mutated breast cancer had a remarkably better 3-month DCR of 81% and 80%, respectively.37,38 Core biopsy and plasma samples from patients with ovarian cancer before and during treatment revealed that PARP inhibitor–ICI treatment combination accentuated interferon-gamma/CXCL9/CXCL10 expression and T lymphocyte tumor infiltration, forming an immunoenhancing microenvironment. 39 The phase I/II TOPACIO trial was a larger study evaluating niraparib in combination with pembrolizumab for patients with ovarian or triple-negative breast cancer with mutated versus wild-type BRCA and revealed an ORR of 25% and DCR of 68%. However, for patients with BRCA mutations, both ORR and DCR were greater at 45% and 73%, respectively.40,41 ORR was also higher at 33% in PD-L1-positive compared to 15% in PD-L1-negative tumors, highlighting the potential utility of PD-L1 as a predictive biomarker for therapeutic response to this combination therapy. 40 Anti-CTLA-4 therapy plus PARPi therapy is another promising approach that warrants further exploration in addition to anti-PD1/PD-L1 therapy. 42

ICI and PARPi have non-overlapping and manageable toxicity profiles. In the MEDIOLA trial, grade 3 or 4 toxicity occurred in less than 10% of patients. 37 The TOPACIO trial also showed that the combination treatment was well tolerated, and there were no new safety signals. 40 Our patient had grade 1 elevated liver function tests, which resolved quickly after a brief discontinuation and could then tolerate reinitiation of the PARPi plus ICI combination.

Of note, there are still several unanswered questions about this treatment combination. The synergy of this combination acting through various mechanisms needs further characterization for routine clinical utility. Our case demonstrated an exceptional response in a type of cancer where current evidence for the use of this combination is limited. The best biomarkers to predict treatment responses are still not clearly elucidated. These are mainly due to the complex interplay of intratumoral and intertumoral heterogeneity and effects from the tumor microenvironment. Biomarkers more specifically predictive of response to the combination of ICI and PARPi may enable us to identify the subset of patients who are likely to derive stronger clinical benefit from the combination approach. Moreover, the mode of tumor sampling (tissue sequencing versus ctDNA), the type of mutation (germline versus somatic), and the PD-L1 expression level could also play roles in determining the treatment responses. 43

Conclusion

We illustrate the case of a patient with metastatic esophageal adenocarcinoma with a germline BRCA2 mutation who only achieved a stable treatment response with the current standard-of-care chemotherapy and ICI combination. Complete resolution of the tumor was then achieved within 4 months of initiation of maintenance PARP inhibitor–ICI therapy. The combination also had a favorable safety profile, consistent with previous studies. This demonstrates that the combination of PARPi and ICI may have synergistic activity through multiple distinct mechanisms in cancers possessing HR deficiencies like BRCA mutations and lead to superior patient outcomes with a tolerable safety profile. Further studies are needed to determine the population that would most benefit from PARP inhibitor–ICI combination treatment strategy to guide therapy in difficult-to-treat cancers and overcome treatment resistance from single-agent PARP inhibitors or ICIs.

Methods

IHC and cytology

Histological evaluation was performed by a pathologist at Saint Luke’s Hospital. IHC of HER2 expression level and HER2 FISH were tested with FDA-cleared rabbit clone 4B5 antibody using Ventana automated platform utilizing an ultra-View Universal Detection Kit at Saint Luke’s Hospital. CPS and TPS were determined using the DAKO PD-L1 22C3 qualitative immunohistochemical assay.

Molecular studies

A tumor sample was obtained from the GE mass via cold forceps biopsies during EGD, then preserved in formalin solution. Tempus xT containing a 648-gene panel DNA sequencing and whole transcriptome RNA sequencing was performed. MSI and TMB were measured using the same panel as well. In addition, the Tempus xF cfDNA panel was used for tumor-normal match and detection of germline alterations, with blood being used as the normal tissue and GEJ mass biopsy as the tumor tissue. Serial ctDNA monitoring was performed during the treatment course using the signatera MRD assay.

The reporting of this study conforms to the CARE guidelines (CARE Checklist – PARPi plus ICI).

Supplemental Material

sj-docx-1-tam-10.1177_17588359241242406 – Supplemental material for Exceptional synergistic response of PARP inhibitor and immune checkpoint inhibitor in esophageal adenocarcinoma with a germline BRCA2 mutation: a case report

Supplemental material, sj-docx-1-tam-10.1177_17588359241242406 for Exceptional synergistic response of PARP inhibitor and immune checkpoint inhibitor in esophageal adenocarcinoma with a germline BRCA2 mutation: a case report by Himil Mahadevia, Ben Ponvilawan, Ammar Al-Obaidi, Jennifer Buckley, Janakiraman Subramanian and Dhruv Bansal in Therapeutic Advances in Medical Oncology

Acknowledgments

We have no acknowledgements to report.

Footnotes

ORCID iD: Himil Mahadevia Inline graphic https://orcid.org/0000-0002-2083-5510

Supplemental material: Supplemental material for this article is available online.

Contributor Information

Himil Mahadevia, Department of Internal Medicine, University of Missouri–Kansas City, Kansas City, MO, USA.

Ben Ponvilawan, Department of Internal Medicine, University of Missouri–Kansas City, Kansas City, MO, USA.

Ammar Al-Obaidi, Department of Hematology and Oncology, University of Missouri–Kansas City, Kansas City, MO, USA.

Jennifer Buckley, Department of Radiology, Saint Luke’s Hospital, Kansas City, MO, USA.

Janakiraman Subramanian, Department of Hematology and Oncology, Inova Schar Cancer Institute, Fairfax, VA, USA.

Dhruv Bansal, Department of Hematology and Oncology, Saint Luke’s Cancer Institute, 4401 Wornall Road, Kansas City, MO 64111, USA.

Declarations

Ethics approval and consent to participate: The patient provided written informed consent for all investigations and treatment, including imaging, histological testing, tumor DNA, and RNA sequencing. The local institutional review board (IRB – at Saint Luke’s Hospital of Kansas City, MO, USA) exempted this study from IRB review.

Consent for publication: The patient provided written informed consent for case report publication.

Author contributions: Himil Mahadevia: Conceptualization; Data curation; Formal analysis; Methodology; Resources; Writing – original draft; Writing – review & editing.

Ben Ponvilawan: Formal analysis; Methodology; Writing – review & editing.

Ammar Al-Obaidi: Methodology; Writing – review & editing.

Jennifer Buckley: Methodology; Writing – review & editing.

Janakiraman Subramanian: Methodology; Writing – review & editing.

Dhruv Bansal: Conceptualization; Methodology; Supervision; Writing – review & editing.

Funding: The authors received no financial support for the research, authorship, and/or publication of this article.

The authors declare that there is no conflict of interest.

Availability of data and materials: The authors declare that data in this study, including the de-identified targeted panel sequencing report, are available through the electronic medical records of Saint Luke’s Hospital of Kansas City. Further inquiries may be directed to the corresponding author.

References

  • 1. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010; 363: 711–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Ansell SM, Lesokhin AM, Borrello I, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med 2015; 372: 311–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Gandhi L, Rodríguez-Abreu D, Gadgeel S, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med 2018; 378: 2078–2092. [DOI] [PubMed] [Google Scholar]
  • 4. Janjigian YY, Shitara K, Moehler M, et al. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): A randomised, open-label, phase 3 trial. Lancet 2021; 398: 27–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Sun J-M, Shen L, Shah MA, et al. Pembrolizumab plus chemotherapy versus chemotherapy alone for first-line treatment of advanced oesophageal cancer (KEYNOTE-590): A randomised, placebo-controlled, phase 3 study. Lancet 2021; 398: 759–771. [DOI] [PubMed] [Google Scholar]
  • 6. Tabernero J, Bang Y-J, Van Cutsem E, et al. KEYNOTE-859: A phase III study of pembrolizumab plus chemotherapy in gastric/gastroesophageal junction adenocarcinoma. Future Oncol 2021; 17: 2847–2855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Moore K, Colombo N, Scambia G, et al. Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med 2018; 379: 2495–2505. [DOI] [PubMed] [Google Scholar]
  • 8. Robson M, Im SA, Senkus E, et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med 2017; 377: 523–533. [DOI] [PubMed] [Google Scholar]
  • 9. Golan T, Hammel P, Reni M, et al. Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N Engl J Med 2019; 381: 317–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. de Bono J, Mateo J, Fizazi K, et al. Olaparib for metastatic castration-resistant prostate cancer. N Engl J Med 2020; 382: 2091–2102. [DOI] [PubMed] [Google Scholar]
  • 11. Rose M, Burgess JT, O’Byrne K, et al. PARP inhibitors: Clinical relevance, mechanisms of action and tumor resistance. Front Cell Dev Biol 2020; 8: 564601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Jin N, Xia Y, Gao Q. Combined PARP inhibitors and small molecular inhibitors in solid tumor treatment (Review). Int J Oncol 2023; 62: 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science 2018; 359: 1350–1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Patel M, Nowsheen S, Maraboyina S, et al. The role of poly(ADP-ribose) polymerase inhibitors in the treatment of cancer and methods to overcome resistance: A review. Cell Biosci 2020; 10: 35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Corrales L, McWhirter SM, Dubensky TW, Jr, et al. The host STING pathway at the interface of cancer and immunity. J Clin Invest 2016; 126: 2404–2411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Yélamos J, Moreno-Lama L, Jimeno J, et al. Immunomodulatory roles of PARP-1 and PARP-2: Impact on PARP-centered cancer therapies. Cancers (Basel) 2020; 12: 392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Jiao S, Xia W, Yamaguchi H, et al. PARP inhibitor upregulates PD-L1 expression and enhances cancer-associated immunosuppression. Clin Cancer Res 2017; 23: 3711–3720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Brown JS, Sundar R, Lopez J. Combining DNA damaging therapeutics with immunotherapy: More haste, less speed. Br J Cancer 2018; 118: 312–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Ding L, Kim HJ, Wang Q, et al. PARP inhibition elicits STING-dependent antitumor immunity in BRCA1-deficient ovarian cancer. Cell Rep 2018; 25: 2972–2980.e2975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Power R, Lowery MA, Reynolds JV, et al. The cancer-immune set point in oesophageal cancer. Front Oncol 2020; 10: 891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Chatterjee N, Walker GC. Mechanisms of DNA damage, repair, and mutagenesis. Environ Mol Mutagen 2017; 58: 235–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Bryant HE, Schultz N, Thomas HD, et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 2005; 434: 913–917. [DOI] [PubMed] [Google Scholar]
  • 23. Helleday T. The underlying mechanism for the PARP and BRCA synthetic lethality: Clearing up the misunderstandings. Mol Oncol 2011; 5: 387–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Liu J, Chen Z, Li Y, et al. PD-1/PD-L1 checkpoint inhibitors in tumor immunotherapy. Front Pharmacol 2021; 12: 731798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. O’Donnell JS, Long GV, Scolyer RA, et al. Resistance to PD1/PDL1 checkpoint inhibition. Cancer Treat Rev 2017; 52: 71–81. [DOI] [PubMed] [Google Scholar]
  • 26. Germano G, Lamba S, Rospo G, et al. Inactivation of DNA repair triggers neoantigen generation and impairs tumour growth. Nature 2017; 552: 116–120. [DOI] [PubMed] [Google Scholar]
  • 27. Zitvogel L, Galluzzi L, Kepp O, et al. Type I interferons in anticancer immunity. Nat Rev Immunol 2015; 15: 405–414. [DOI] [PubMed] [Google Scholar]
  • 28. Li CW, Lim SO, Xia W, et al. Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity. Nat Commun 2016; 7: 12632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Mouw KW, Konstantinopoulos PA. From checkpoint to checkpoint: DNA damage ATR/Chk1 checkpoint signalling elicits PD-L1 immune checkpoint activation. Br J Cancer 2018; 118: 933–935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Doki Y, Ajani JA, Kato K, et al. Nivolumab combination therapy in advanced esophageal squamous-cell carcinoma. New Engl J Med 2022; 386: 449–462. [DOI] [PubMed] [Google Scholar]
  • 31. Kojima T, Shah MA, Muro K, et al. Randomized phase III KEYNOTE-181 study of pembrolizumab versus chemotherapy in advanced esophageal cancer. J Clin Oncol 2020; 38: 4138–4148. [DOI] [PubMed] [Google Scholar]
  • 32. Kato K, Cho BC, Takahashi M, et al. Nivolumab versus chemotherapy in patients with advanced oesophageal squamous cell carcinoma refractory or intolerant to previous chemotherapy (ATTRACTION-3): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2019; 20: 1506–1517. [DOI] [PubMed] [Google Scholar]
  • 33. Bang YJ, Xu RH, Chin K, et al. Olaparib in combination with paclitaxel in patients with advanced gastric cancer who have progressed following first-line therapy (GOLD): A double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol 2017; 18: 1637–1651. [DOI] [PubMed] [Google Scholar]
  • 34. Huang KL, Mashl RJ, Wu Y, et al. Pathogenic germline variants in 10,389 adult cancers. Cell 2018; 173: 355–370.e314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Bang Y-J, Kaufman B, Geva R, et al. An open-label, phase II basket study of olaparib and durvalumab (MEDIOLA): Results in patients with relapsed gastric cancer. J Clin Oncol 2019; 37: 140–140. [Google Scholar]
  • 36. Thomas A, Vilimas R, Trindade C, et al. Durvalumab in combination with olaparib in patients with relapsed SCLC: Results from a phase II study. J Thorac Oncol 2019; 14: 1447–1457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Domchek SM, Postel-Vinay S, Im SA, et al. Olaparib and durvalumab in patients with germline BRCA-mutated metastatic breast cancer (MEDIOLA): An open-label, multicentre, phase 1/2, basket study. Lancet Oncol 2020; 21: 1155–1164. [DOI] [PubMed] [Google Scholar]
  • 38. Lee J-M, Annunziata C, Houston N, et al. A phase II study of durvalumab, a PD-L1 inhibitor and olaparib in recurrent ovarian cancer (OvCa). Ann Oncol 2018; 29: viii334. [Google Scholar]
  • 39. Lampert EJ, Zimmer A, Padget M, et al. Combination of PARP inhibitor olaparib, and PD-L1 inhibitor durvalumab, in recurrent ovarian cancer: A proof-of-concept phase II study. Clin Cancer Res 2020; 26: 4268–4279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Konstantinopoulos PA, Waggoner S, Vidal GA, et al. Single-arm phases 1 and 2 trial of niraparib in combination with pembrolizumab in patients with recurrent platinum-resistant ovarian carcinoma. JAMA Oncol 2019; 5: 1141–1149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Vinayak S, Tolaney SM, Schwartzberg L, et al. Open-label clinical trial of niraparib combined with pembrolizumab for treatment of advanced or metastatic triple-negative breast cancer. JAMA Oncol 2019; 5: 1132–1140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Adams SF, Rixe O, Lee J-H, et al. Phase I study combining olaparib and tremelimumab for the treatment of women with BRCA-deficient recurrent ovarian cancer. J Clin Oncol 2017; 35: e17052–e17052. [Google Scholar]
  • 43. Peyraud F, Italiano A. Combined PARP inhibition and immune checkpoint therapy in solid tumors. Cancers (Basel) 2020; 12: 1502. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

sj-docx-1-tam-10.1177_17588359241242406 – Supplemental material for Exceptional synergistic response of PARP inhibitor and immune checkpoint inhibitor in esophageal adenocarcinoma with a germline BRCA2 mutation: a case report

Supplemental material, sj-docx-1-tam-10.1177_17588359241242406 for Exceptional synergistic response of PARP inhibitor and immune checkpoint inhibitor in esophageal adenocarcinoma with a germline BRCA2 mutation: a case report by Himil Mahadevia, Ben Ponvilawan, Ammar Al-Obaidi, Jennifer Buckley, Janakiraman Subramanian and Dhruv Bansal in Therapeutic Advances in Medical Oncology


Articles from Therapeutic Advances in Medical Oncology are provided here courtesy of SAGE Publications

RESOURCES