Trends in Cancer
Volume 7, Issue 10, October 2021, Pages 958-970
Journal home page for Trends in Cancer

Review
Development of poly(ADP-ribose) polymerase inhibitor and immunotherapy combinations: progress, pitfalls, and promises

https://doi.org/10.1016/j.trecan.2021.05.004Get rights and content

Highlights

  • Poly(ADP-ribose) polymerase inhibitors (PARPi) are approved by the FDA as monotherapy for ovarian, breast, and pancreatic tumors harboring BRCA1/2 mutations, as well as castration-resistant prostate cancers with homologous recombination repair (HRR) mutations.

  • Immune checkpoint blockade (ICB) – through the use of programmed cell death 1/programmed cell death ligand 1 (PD-1/PD-L1) inhibitors – has demonstrated durable responses in different solid tumors. Several PD-1/PD-L1 inhibitors are FDA approved for non-small cell lung cancer, metastatic urothelial carcinoma, and metastatic cutaneous squamous cell carcinoma.

  • Preclinical data support a synergistic relationship between PARPi and ICB; PARPi-mediated DNA damage modulates the tumor immune microenvironment though different molecular and cellular mechanisms, such as increasing genomic instability, immune pathway activation, and PD-L1 expression on cancer cells; all of which may increase responsiveness to ICB.

  • Early data from a Phase I trial suggested that the combination of PARPi and anti-PD-1/PD-L1 may be beneficial in patients regardless of mutational status or even in the case of platinum sensitivity. Thus far, it has been shown that combining PARPi with ICB may be a safe and well tolerated strategy, with the potential to improve survival outcomes in a broad population of patients with BRCA1/2 mutations.

The efficacy of poly(ADP-ribose) polymerase inhibitors (PARPi) is restricted by inevitable drug resistance, while their use in combination with chemotherapy and targeted agents is commonly associated with dose-limiting toxicities. Immune checkpoint blockade (ICB) has demonstrated durable responses in different solid tumors and is well-established across multiple cancers. Despite this, single agent activity is limited to a minority of patients and drug resistance remains an issue. Building on the monotherapy success of both drug classes, combining PARPi with ICB may be a safe and well-tolerated strategy with the potential to improve survival outcomes. In this review, we present the preclinical, translational, and clinical data supporting the combination of DNA damage response (DDR) and ICB as well as consider important questions to be addressed with future research.

Introduction

The field of oncology drug development has seen substantial progress in the treatment of different cancers over the past decade. Inhibitors of key DDR mechanisms, specifically against PARP, have proven to be effective and are now approved as single-agent therapies in breast, ovarian, pancreatic, and castration-resistant prostate cancers (CRPC) [1]. ICB agents targeting cytotoxic T lymphocyte antigen (CTLA)-4 and programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1) have also emerged as effective strategies to improve survival outcomes in various solid tumors [2]. However, clinical benefit for both classes of drugs is limited by innate and acquired resistance and/or drug-related toxicities. Preclinical studies have supported the mechanistic rationale for the combination of PARPi and ICB as a therapeutic strategy for patients with different solid tumors [3].

In this review, we discuss the preclinical, translational, and clinical studies supporting the combination of PARPi and ICB therapies as well as consider important questions to be addressed with future research.

Section snippets

PARP and synthetic lethality

The PARP family of enzymes consists of 17 proteins that play important roles in the DDR pathways for base excision repair (BER) and single-strand break repair (SSB) [4., 5., 6.]. PARP1, the most widely studied protein of this family, is composed of a common catalytic domain, which generates poly(ADP-ribose) chains upon binding to SSB through a process known as auto-PARylation [4,7,8]. These PARP complexes promote the downstream recruitment of SSB repair proteins and lead to the chromatin

Clinical activity of PARPi

Four randomized Phase III trials – SOLO-1 (NCT01844986i), PAOLA-1/ENGOT-OV25 (NCT02477644ii), PRIMA/ENGOT-OV26 (NCT02655016iii), and VELIA/GOG-3005 (NCT02470585iv) – have demonstrated clinically significant progression-free survival (PFS) in newly diagnosed ovarian cancer patients undergoing PARPi treatment. Olaparib and rucaparib have already received FDA approval for the treatment of BRCA1/2 mutated recurrent, platinum-sensitive ovarian cancer. Olaparib, niraparib, and rucaparib have been

Immuno-oncology and ICB

Immuno-oncology (IO) therapy targets the complex adaptive changes in the tumor microenvironment upon antigen recognition [33]. An interesting IO target is CTLA-4, a co-inhibitory molecule found on the cell surface of regulatory T cells that interacts with antigen-presenting cells (APC) [34]. In conjunction with the downregulation of co-stimulatory CD80 and CD86 cells, CTLA-4 suppresses T cell function and mitigates the amplitude of the cytotoxic T cell response [33,34]. Murine models have

Combining PARP inhibition and immunotherapy: a rational approach

Despite the approval of different PD-1/PD-L1 agents for the treatment of multiple cancer types, only a minority of patients will benefit from a monotherapeutic approach [36,41]. This is exemplified in gynecological cancers, where the use of PD-1/PD-L1 agents has demonstrated modest 10–15% response rates in ovarian, cervical, and endometrial cancers [44,45]. Recently, we have observed a shift towards developing rational combinatorial strategies to enhance the efficacy of ICB, including

Patient selection

To maximize the number of patients who may benefit from combined PARP and immune checkpoint inhibition, we must optimize patient selection through the use of response and resistance predictive biomarkers [9]. While tumors with other known mechanisms of HR deficiency or ‘BRCAness’ may also benefit from exploiting the synthetic lethality relationship with PARPi beyond BRCA1/2 mutations, empirically targeting other HRR alterations has yet to show a comparable level of clinical benefit [1]. What

Concluding remarks

Despite FDA approval for use of PARPi in HR-deficient CRPC and BRCA1/2-mutated ovarian, breast, and pancreatic tumors, the efficacy of single agent use and combination therapy has been limited by drug resistance and toxicity. Similarly, durable responses with ICB single therapy have been limited to a minority of patients and tumor types. Robust preclinical data have suggested activation of the cGAS-STING pathway by PARP inhibition, shifting the tumor immune microenvironment from one of

Acknowledgments

M.M.P. is supported by the National Institutes of Health (T32 CA101642/CA/NCI NIH HHS/United States). N.Y.L.N. is supported by the National Medical Research Council, Singapore (MOH-FLWSHP19may-0006). D.S.P. T. is supported by the National Medical Research Council, Singapore (CSAINV16may008), and has received charitable research funding from the Pangestu Family Foundation Gynaecological Cancer Research Fund. G.P. is supported by MD Anderson Cancer Center Support grant (NCI CA016672), the

Declaration of interests

N.Y.L.N. received honoraria from AstraZeneca and Janssen. D.S.P.T. received research funding from AstraZeneca, Bayer, and Karyopharm and honoraria from AstraZeneca, MSD, Tessa Therapeutics, Novartis, Bayer, and Genmab. T.A.Y. received research funding (paid to his institution) from Artios, AstraZeneca, Bayer, Clovis, Constellation, Cyteir, Eli Lilly, EMD Serono, Forbius, F-Star, GlaxoSmithKline, Genentech, ImmuneSensor, Ipsen, Jounce, Karyopharm, Kyowa, Merck, Novartis, Pfizer, Ribon

References (77)

  • R. Coleman

    Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double-blind, placebo-controlled, phase 3 trial

    Lancet

    (2017)
  • Y. Drew

    Phase II study of olaparib + durvalumab (MEDIOLA): Updated results in germline BRCA-mutated platinum-sensitive relapsed (PSR) ovarian cancer (OC)

    Ann. Oncol.

    (2019)
  • S. Domchek

    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)
  • J.S. Brown

    Targeting DNA repair in cancer: beyond PARP inhibitors

    Cancer Discov.

    (2017)
  • D. Pardoll

    The blockade of immune checkpoints in cancer immunotherapy

    Nat. Rev. Cancer

    (2012)
  • J. Shen

    PARPi triggers the STING-dependent immune response and enhances the therapeutic efficacy of immune checkpoint blockade independent of BRCAness

    Cancer Res.

    (2019)
  • A. Cerrato

    Use of poly ADP-ribose polymerase [PARP] inhibitors in cancer cells bearing DDR defects: the rationale for their inclusion in the clinic

    J. Exp. Clin. Cancer Res.

    (2016)
  • M. Satoh et al.

    Role of poly(ADP-ribose) formation in DNA repair

    Nature

    (1992)
  • S. Howard

    DNA damage response factors from diverse pathways, including DNA crosslink repair, mediate alternative end joining

    PLoS Genet.

    (2015)
  • E. Huarte

    Depletion of dendritic cells delays ovarian cancer progression by boosting antitumor immunity

    Cancer Res.

    (2008)
  • C.J. Lord et al.

    PARP inhibitors: the first synthetic lethal targeted therapy

    Science (80- )

    (2017)
  • J. Murai

    Trapping of PARP1 and PARP2 by clinical PARP inhibitors

    Cancer Res.

    (2012)
  • M. Rouleau

    PARP inhibition: PARP1 and beyond

    Nat. Rev. Cacner

    (2010)
  • A. Chaudhuri et al.

    The multifaceted roles of PARP1 in DNA repair and chromatin remodelling Arnab

    Nat. Rev. Mol. Cell Biol.

    (2017)
  • M.E. Moynahan

    Homology-directed DNA repair, mitomycin-C resistance, and chromosome stability is restored with correction of a Brca1 mutation

    Cancer Res.

    (2001)
  • P. Fong

    Inhibition of Poly(ADP-Ribose) polymerase in tumors from BRCA mutation carriers

    N. Engl. J. Med.

    (2009)
  • L. Pearl

    Therapeutic opportunities within the DNA damage response

    Nat. Rev. Cancer

    (2015)
  • H. Farmer

    Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy

    Nature

    (2005)
  • H. Bryant

    Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase

    Nature

    (2005)
  • Y. Shen

    BMN 673, a novel and highly potent PARP1 / 2 inhibitor for the treatment of human cancers with DNA repair deficiency

    (2019)
  • J. Murai

    Stereospecific PARP trapping by BMN 673 and comparison with olaparib and rucaparib

    (2015)
  • Y. Pommier

    Laying a trap to kill cancer cells: PARP inhibitors and their mechanisms of action

    Sci. Transl. Med.

    (2016)
  • J. Murai

    Targeting DNA repair adn replication stress in the treatment of ovarian cancer

    Int. J. Clin. Oncol.

    (2017)
  • M. Patel

    The role of poly(ADP-ribose) polymerase inhibitors in the treatment of cancer and methods to overcome resistance: a review

    Cell Biosci.

    (2020)
  • M. Mirza

    The forefront of ovarian cancer therapy: update on PARP inhibitors

    Ann. Oncol.

    (2020)
  • R.L. Coleman

    Veliparib with first-line chemotherapy and as maintenance therapy in ovarian cancer

    N. Engl. J. Med.

    (2019)
  • K. Moore

    Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer

    N. Engl. J. Med.

    (2018)
  • M.R. Mirza

    Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer

    N. Engl. J. Med.

    (2016)
  • Cited by (18)

    • PARP1 controls the transcription of CD24 by ADP-ribosylating the RNA helicase DDX5 in pancreatic cancer

      2023, International Journal of Biochemistry and Cell Biology
      Citation Excerpt :

      These findings directly link PARP1 to the immune system and reveal that PARP1 is involved in the tumor immune microenvironment through its PARylation activity. PARP blockade has been shown to modulate the tumor microenvironment at the molecular and cellular levels by means such as increasing immune pathway activation and PD-L1 expression on cancer cells, thus potentially affecting immune checkpoint inhibitor responsiveness (Hagiwara et al., 2022, Pham et al., 2021). Combined use of anti-PD-L1 or anti-PD-1 therapy sensitized tumors to PARP-targeted therapy and enhanced antitumor effects in several cancers, such as advanced non-small cell lung cancer, triple-negative breast cancer and ovarian cancer (Konstantinopoulos et al., 2019, Ramalingam et al., 2022, Vinayak et al., 2019).

    • Targeting ATR in patients with cancer

      2024, Nature Reviews Clinical Oncology
    View all citing articles on Scopus
    View full text