1932

Abstract

Innate immunity and the DNA damage response (DDR) pathway are inextricably linked. Within the DDR, ataxia telangiectasia and Rad3-related (ATR) is a key kinase responsible for sensing replication stress and facilitating DNA repair through checkpoint activation, cell cycle arrest, and promotion of fork recovery. Recent studies have shed light on the immunomodulatory role of the ATR-CHK1 pathway in the tumor microenvironment and the specific effects of ATR inhibition in stimulating an innate immune response. With several potent and selective ATR inhibitors in developmental pipelines, the combination of dual ATR and PD-(L)1 blockade has attracted increasing interest in cancer therapy. In this review, we summarize the clinical and preclinical data supporting the combined inhibition of ATR and PD-(L)1, discuss the potential challenges surrounding this approach, and highlight biomarkers relevant for selected patients who are most likely to benefit from the blockade of these two checkpoints.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-042320-025136
2022-01-27
2024-05-13
Loading full text...

Full text loading...

/deliver/fulltext/med/73/1/annurev-med-042320-025136.html?itemId=/content/journals/10.1146/annurev-med-042320-025136&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Branzei D, Foiani M. 2010. Maintaining genome stability at the replication fork. Nat. Rev. Mol. Cell Biol. 11:208–19
    [Google Scholar]
  2. 2. 
    Jackson SP, Bartek J 2009. The DNA-damage response in human biology and disease. Nature 461:1071–78
    [Google Scholar]
  3. 3. 
    Matsuoka S, Ballif BA, Smogorzewska A et al. 2007. ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA damage. Science 316:1160–66
    [Google Scholar]
  4. 4. 
    Byun TS, Pacek M, Yee MC et al. 2005. Functional uncoupling of MCM helicase and DNA polymerase activities activates the ATR-dependent checkpoint. Genes Dev 19:1040–52
    [Google Scholar]
  5. 5. 
    Zou L, Elledge SJ. 2003. Sensing DNA damage through ATRIP recognition of RPA-ssDNA complexes. Science 300:1542–48
    [Google Scholar]
  6. 6. 
    Dobbelstein M, Sorensen CS. 2015. Exploiting replicative stress to treat cancer. Nat. Rev. Drug Discov. 14:405–23
    [Google Scholar]
  7. 7. 
    Liptay M, Barbosa JS, Rottenberg S. 2020. Replication fork remodeling and therapy escape in DNA damage response–deficient cancers. Front. Oncol. 10:670
    [Google Scholar]
  8. 8. 
    Matos DA, Zhang JM, Ouyang J et al. 2020. ATR protects the genome against R loops through a MUS81-triggered feedback loop. Mol. Cell 77:514–27.e4
    [Google Scholar]
  9. 9. 
    Santos-Pereira JM, Aguilera A. 2015. R loops: new modulators of genome dynamics and function. Nat. Rev. Genet. 16:583–97
    [Google Scholar]
  10. 10. 
    Bahassi EM, Ovesen JL, Riesenberg AL et al. 2008. The checkpoint kinases Chk1 and Chk2 regulate the functional associations between hBRCA2 and Rad51 in response to DNA damage. Oncogene 27:3977–85
    [Google Scholar]
  11. 11. 
    Sorensen CS, Hansen LT, Dziegielewski J et al. 2005. The cell-cycle checkpoint kinase Chk1 is required for mammalian homologous recombination repair. Nat. Cell Biol. 7:195–201
    [Google Scholar]
  12. 12. 
    Berti M, Vindigni A 2016. Replication stress: getting back on track. Nat. Struct. Mol. Biol. 23:103–9
    [Google Scholar]
  13. 13. 
    Negrini S, Gorgoulis VG, Halazonetis TD. 2010. Genomic instability—an evolving hallmark of cancer. Nat. Rev. Mol. Cell Biol. 11:220–28
    [Google Scholar]
  14. 14. 
    Gomes LR, Rocha CRR, Martins DJ et al. 2019. ATR mediates cisplatin resistance in 3D-cultured breast cancer cells via translesion DNA synthesis modulation. Cell Death Dis 10:459
    [Google Scholar]
  15. 15. 
    Ashley AK, Kemp CJ. 2018. DNA-PK, ATM, and ATR: PIKKing on p53. Cell Cycle 17:275–76
    [Google Scholar]
  16. 16. 
    Williamson CT, Miller R, Pemberton HN et al. 2016. ATR inhibitors as a synthetic lethal therapy for tumours deficient in ARID1A. Nat. Commun. 7:13837
    [Google Scholar]
  17. 17. 
    Vendetti FP, Lau A, Schamus S et al. 2015. The orally active and bioavailable ATR kinase inhibitor AZD6738 potentiates the anti-tumor effects of cisplatin to resolve ATM-deficient non-small cell lung cancer in vivo. Oncotarget 6:44289–305
    [Google Scholar]
  18. 18. 
    Gilad O, Nabet BY, Ragland RL et al. 2010. Combining ATR suppression with oncogenic Ras synergistically increases genomic instability, causing synthetic lethality or tumorigenesis in a dosage-dependent manner. Cancer Res 70:9693–702
    [Google Scholar]
  19. 19. 
    Kok YP, Guerrero Llobet S, Schoonen PM et al. 2020. Overexpression of cyclin E1 or Cdc25A leads to replication stress, mitotic aberrancies, and increased sensitivity to replication checkpoint inhibitors. Oncogenesis 9:88
    [Google Scholar]
  20. 20. 
    Savva C, De Souza K, Ali R et al. 2019. Clinicopathological significance of ataxia telangiectasia-mutated (ATM) kinase and ataxia telangiectasia-mutated and Rad3-related (ATR) kinase in MYC overexpressed breast cancers. Breast Cancer Res. Treat. 175:105–15
    [Google Scholar]
  21. 21. 
    Nguyen HD, Zou L, Graubert TA. 2019. Targeting R-loop-associated ATR response in myelodysplastic syndrome. Oncotarget 10:2581–82
    [Google Scholar]
  22. 22. 
    Dvinge H, Kim E, Abdel-Wahab O, Bradley RK 2016. RNA splicing factors as oncoproteins and tumour suppressors. Nat. Rev. Cancer 16:413–30
    [Google Scholar]
  23. 23. 
    Nguyen HD, Yadav T, Giri S et al. 2017. Functions of replication protein A as a sensor of R loops and a regulator of RNaseH1. Mol. Cell 65:832–47.e4
    [Google Scholar]
  24. 24. 
    Nguyen HD, Leong WY, Li W et al. 2018. Spliceosome mutations induce R loop–associated sensitivity to ATR inhibition in myelodysplastic syndromes. Cancer Res 78:5363–74
    [Google Scholar]
  25. 25. 
    Dillon M, Guevara J, Mohammed K et al. 2019. A phase I study of ATR inhibitor, AZD6738, as monotherapy in advanced solid tumours (PATRIOT part A, B). Ann. Oncol. 30:5165–66
    [Google Scholar]
  26. 26. 
    Yap TA, O'Carrigan B, Penney MS et al. 2020. Phase I trial of first-in-class ATR inhibitor M6620 (VX-970) as monotherapy or in combination with carboplatin in patients with advanced solid tumors. J. Clin. Oncol. 38:3195–204
    [Google Scholar]
  27. 27. 
    Hall AB, Newsome D, Wang Y et al. 2014. Potentiation of tumor responses to DNA damaging therapy by the selective ATR inhibitor VX-970. Oncotarget 5:5674–85
    [Google Scholar]
  28. 28. 
    Combes E, Andrade AF, Tosi D et al. 2019. Inhibition of ataxia-telangiectasia mutated and RAD3-related (ATR) overcomes oxaliplatin resistance and promotes antitumor immunity in colorectal cancer. Cancer Res 79:2933–46
    [Google Scholar]
  29. 29. 
    Ngoi NY, Sundararajan V, Tan DS. 2020. Exploiting replicative stress in gynecological cancers as a therapeutic strategy. Int. J. Gynecol. Cancer 30:1224–38
    [Google Scholar]
  30. 30. 
    Wallez Y, Dunlop CR, Johnson TI et al. 2018. The ATR inhibitor AZD6738 synergizes with gemcitabine in vitro and in vivo to induce pancreatic ductal adenocarcinoma regression. Mol. Cancer Ther. 17:1670–82
    [Google Scholar]
  31. 31. 
    Konstantinopoulos PA, Cheng SC, Wahner Hendrickson AE et al. 2020. Berzosertib plus gemcitabine versus gemcitabine alone in platinum-resistant high-grade serous ovarian cancer: a multicentre, open-label, randomised, phase 2 trial. Lancet Oncol 21:957–68
    [Google Scholar]
  32. 32. 
    Gasser S, Orsulic S, Brown EJ, Raulet DH. 2005. The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 436:1186–90
    [Google Scholar]
  33. 33. 
    Sato H, Niimi A, Yasuhara T et al. 2017. DNA double-strand break repair pathway regulates PD-L1 expression in cancer cells. Nat. Commun. 8:1751
    [Google Scholar]
  34. 34. 
    Chen CF, Ruiz-Vega R, Vasudeva P et al. 2017. ATR mutations promote the growth of melanoma tumors by modulating the immune microenvironment. Cell Rep 18:2331–42
    [Google Scholar]
  35. 35. 
    Lee J, Kim ST, Smith S et al. 2020. Results from a phase I, open-label study of ceralasertib (AZD6738), a novel DNA damage repair agent, in combination with weekly paclitaxel in refractory cancer (NCT02630199). J. Clin. Oncol. 38:3503
    [Google Scholar]
  36. 36. 
    Reck M, Rodríguez-Abreu D, Robinson AG et al. 2016. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N. Engl. J. Med. 375:1823–33
    [Google Scholar]
  37. 37. 
    Hellmann MD, Paz-Ares L, Bernabe Caro R et al. 2019. Nivolumab plus ipilimumab in advanced non-small-cell lung cancer. N. Engl. J. Med. 381:2020–31
    [Google Scholar]
  38. 38. 
    Hellmann MD, Ciuleanu TE, Pluzanski A et al. 2018. Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N. Engl. J. Med. 378:2093–104
    [Google Scholar]
  39. 39. 
    Baas P, Scherpereel A, Nowak AK et al. 2021. First-line nivolumab plus ipilimumab in unresectable malignant pleural mesothelioma (CheckMate 743): a multicentre, randomised, open-label, phase 3 trial. Lancet 397:375–86
    [Google Scholar]
  40. 40. 
    Motzer RJ, Tannir NM, McDermott DF et al. 2018. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N. Engl. J. Med. 378:1277–90
    [Google Scholar]
  41. 41. 
    El-Khoueiry AB, Sangro B, Yau T et al. 2017. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 389:2492–502
    [Google Scholar]
  42. 42. 
    Robert C, Schachter J, Long GV et al. 2015. Pembrolizumab versus ipilimumab in advanced melanoma. N. Engl. J. Med. 372:2521–32
    [Google Scholar]
  43. 43. 
    Balar AV, Castellano D, O'Donnell PH et al. 2017. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol 18:1483–92
    [Google Scholar]
  44. 44. 
    Agata Y, Kawasaki A, Nishimura H et al. 1996. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int. Immunol. 8:765–72
    [Google Scholar]
  45. 45. 
    Arasanz H, Gato-Cañas M, Zuazo M et al. 2017. PD1 signal transduction pathways in T cells. Oncotarget 8:51936–45
    [Google Scholar]
  46. 46. 
    Waldman AD, Fritz JM, Lenardo MJ. 2020. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat. Rev. Immunol. 20:651–68
    [Google Scholar]
  47. 47. 
    Bonaventura P, Shekarian T, Alcazer V et al. 2019. Cold tumors: a therapeutic challenge for immunotherapy. Front. Immunol. 10:168
    [Google Scholar]
  48. 48. 
    Barber GN. 2015. STING: infection, inflammation and cancer. Nat. Rev. Immunol. 15:760–70
    [Google Scholar]
  49. 49. 
    Chatzinikolaou G, Karakasilioti I, Garinis GA. 2014. DNA damage and innate immunity: links and trade-offs. Trends Immunol 35:429–35
    [Google Scholar]
  50. 50. 
    Mouw KW, Goldberg MS, Konstantinopoulos PA, D'Andrea AD. 2017. DNA damage and repair biomarkers of immunotherapy response. Cancer Discov 7:675–93
    [Google Scholar]
  51. 51. 
    Ablasser A, Goldeck M, Cavlar T et al. 2013. cGAS produces a 2′–5′-linked cyclic dinucleotide second messenger that activates STING. Nature 498:380–84
    [Google Scholar]
  52. 52. 
    Chen Q, Sun L, Chen ZJ 2016. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat. Immunol. 17:1142–49
    [Google Scholar]
  53. 53. 
    Parkes EE, Walker SM, Taggart LE et al. 2017. Activation of STING-dependent innate immune signaling by S-phase-specific DNA damage in breast cancer. J. Natl. Cancer Inst. 109:djw199
    [Google Scholar]
  54. 54. 
    Pilié PG, Tang Z, Park S et al. 2019. Inhibitors of ataxia-telangiectasia related (ATR) protein lead to innate immune pathway activation and enhanced response to immune therapy Work. Pap., MD Anderson Cancer Cent Houston, TX: https://www.pcf.org/wp-content/uploads/2019/09/Pilie_Patrick.pdf
  55. 55. 
    Yap TA, Tan DSP, Terbuch A et al. 2020. First-in-human trial of the oral ataxia telangiectasia and RAD3-related (ATR) inhibitor BAY 1895344 in patients with advanced solid tumors. Cancer Discov 11:80–91
    [Google Scholar]
  56. 56. 
    Alimzhanov M, Soulard P, Zimmermann A et al. 2019. ATR inhibitor M6620 enhances anti-tumor efficacy of the combination of the anti-PD-L1 antibody avelumab with platinum-based chemotherapy. Cancer Res 79:2269 Abstr.)
    [Google Scholar]
  57. 57. 
    Sheng H, Huang Y, Xiao Y et al. 2020. ATR inhibitor AZD6738 enhances the antitumor activity of radiotherapy and immune checkpoint inhibitors by potentiating the tumor immune microenvironment in hepatocellular carcinoma. J. Immunother. Cancer 8:e000340
    [Google Scholar]
  58. 58. 
    Vendetti FP, Karukonda P, Clump DA et al. 2018. ATR kinase inhibitor AZD6738 potentiates CD8+ T cell–dependent antitumor activity following radiation. J. Clin. Investig. 128:3926–40
    [Google Scholar]
  59. 59. 
    Dillon MT, Bergerhoff KF, Pedersen M et al. 2019. ATR inhibition potentiates the radiation-induced inflammatory tumor microenvironment. Clin. Cancer Res. 25:3392–403
    [Google Scholar]
  60. 60. 
    Chen J, Harding SM, Natesan R et al. 2020. Cell cycle checkpoints cooperate to suppress DNA- and RNA-associated molecular pattern recognition and anti-tumor immune responses. Cell Rep 32:108080
    [Google Scholar]
  61. 61. 
    Harding SM, Benci JL, Irianto J et al. 2017. Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature 548:466–70
    [Google Scholar]
  62. 62. 
    Kakoti S, Sato H, Laskar S et al. 2020. DNA repair and signaling in immune-related cancer therapy. Front. Mol. Biosci 7:205
    [Google Scholar]
  63. 63. 
    Besse B, Awad M, Forde P et al. 2021. OA07.08 HUDSON: an open-label, multi-drug, biomarker-directed, phase II platform study in patients with NSCLC, who progressed on anti-PD(L)1 therapy. J. Thorac. Oncol. 16:S118–19
    [Google Scholar]
  64. 64. 
    Li A, Yi M, Qin S et al. 2019. Prospects for combining immune checkpoint blockade with PARP inhibition. J. Hematol. Oncol. 12:98
    [Google Scholar]
  65. 65. 
    Bryant HE, Schultz N, Thomas HD et al. 2005. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 434:913–17
    [Google Scholar]
  66. 66. 
    Farmer H, McCabe N, Lord CJ et al. 2005. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434:917–21
    [Google Scholar]
  67. 67. 
    Murai J, Huang SY, Das BB et al. 2012. Trapping of PARP1 and PARP2 by clinical PARP inhibitors. Cancer Res 72:5588–99
    [Google Scholar]
  68. 68. 
    Maya-Mendoza A, Moudry P, Merchut-Maya JM et al. 2018. High speed of fork progression induces DNA replication stress and genomic instability. Nature 559:279–84
    [Google Scholar]
  69. 69. 
    Vikas P, Borcherding N, Chennamadhavuni A, Garje R 2020. Therapeutic potential of combining PARP inhibitor and immunotherapy in solid tumors. Front. Oncol. 10:570
    [Google Scholar]
  70. 70. 
    Higuchi T, Flies DB, Marjon NA et al. 2015. CTLA-4 blockade synergizes therapeutically with PARP inhibition in BRCA1-deficient ovarian cancer. Cancer Immunol. Res. 3:1257–68
    [Google Scholar]
  71. 71. 
    Shen J, Zhao W, Ju Z et al. 2019. PARPi triggers the STING-dependent immune response and enhances the therapeutic efficacy of immune checkpoint blockade independent of BRCAness. Cancer Res 79:311–19
    [Google Scholar]
  72. 72. 
    Lampert EJ, Zimmer A, Padget M et al. 2020. Combination of PARP inhibitor olaparib, and PD-L1 inhibitor durvalumab, in recurrent ovarian cancer: a proof-of-concept phase II study. Clin. Cancer Res. 26:4268–79
    [Google Scholar]
  73. 73. 
    Bang Y-J, Kaufman B, Geva R et al. 2019. An open-label, phase II basket study of olaparib and durvalumab (MEDIOLA): results in patients with relapsed gastric cancer. J. Clin. Oncol. 37:140 Abstr.)
    [Google Scholar]
  74. 74. 
    Domchek SM, Postel-Vinay S, Im SA et al. 2020. Olaparib and durvalumab in patients with germline BRCA-mutated metastatic breast cancer (MEDIOLA): an open-label, multicentre, phase 1/2, basket study. Lancet Oncol 21:1155–64
    [Google Scholar]
  75. 75. 
    Schoonen PM, Talens F, Stok C et al. 2017. Progression through mitosis promotes PARP inhibitor–induced cytotoxicity in homologous recombination–deficient cancer cells. Nat. Commun. 8:15981
    [Google Scholar]
  76. 76. 
    Choi M, Kipps T, Kurzrock R. 2016. ATM mutations in cancer: therapeutic implications. Mol. Cancer Ther. 15:1781–91
    [Google Scholar]
  77. 77. 
    Davis AA, Patel VG. 2019. The role of PD-L1 expression as a predictive biomarker: an analysis of all US Food and Drug Administration (FDA) approvals of immune checkpoint inhibitors. J. Immunother. Cancer 7:278
    [Google Scholar]
  78. 78. 
    FDA (US Food Drug Adm.) 2020. FDA approves pembrolizumab for adults and children with TMB-H solid tumors News Release, June 16, FDA Washington, DC:
  79. 79. 
    Buisson R, Boisvert JL, Benes CH, Zou L. 2015. Distinct but concerted roles of ATR, DNA-PK, and Chk1 in countering replication stress during S phase. Mol. Cell 59:1011–24
    [Google Scholar]
  80. 80. 
    Hill SJ, Decker B, Roberts EA et al. 2018. Prediction of DNA repair inhibitor response in short-term patient-derived ovarian cancer organoids. Cancer Discov 8:1404–21
    [Google Scholar]
  81. 81. 
    Chuah S, Chew V. 2020. High-dimensional immune-profiling in cancer: implications for immunotherapy. J. Immunother. Cancer 8:e000363
    [Google Scholar]
  82. 82. 
    Karzai F, VanderWeele D, Madan RA et al. 2018. Activity of durvalumab plus olaparib in metastatic castration-resistant prostate cancer in men with and without DNA damage repair mutations. J. Immunother. Cancer 6:141
    [Google Scholar]
  83. 83. 
    Färkkilä A, Gulhan DC, Casado J et al. 2020. Immunogenomic profiling determines responses to combined PARP and PD-1 inhibition in ovarian cancer. Nat. Commun. 11:1459
    [Google Scholar]
  84. 84. 
    Thomas A, Vilimas R, Trindade C et al. 2019. Durvalumab in combination with olaparib in patients with relapsed SCLC: results from a phase II study. J. Thorac. Oncol. 14:1447–57
    [Google Scholar]
  85. 85. 
    Mouw KW, Konstantinopoulos PA. 2018. From checkpoint to checkpoint: DNA damage ATR/Chk1 checkpoint signalling elicits PD-L1 immune checkpoint activation. Br. J. Cancer 118:933–35
    [Google Scholar]
  86. 86. 
    Kwon J, Bakhoum SF. 2020. The cytosolic DNA-sensing cGAS-STING pathway in cancer. Cancer Discov 10:26–39
    [Google Scholar]
  87. 87. 
    Krebs MG, Lopez J, El-Khoueiry A et al. 2018. Phase I study of AZD6738, an inhibitor of ataxia telangiectasia Rad3-related (ATR), in combination with olaparib or durvalumab in patients (pts) with advanced solid cancers. Cancer Res 78:CT026 Abstr.)
    [Google Scholar]
/content/journals/10.1146/annurev-med-042320-025136
Loading
/content/journals/10.1146/annurev-med-042320-025136
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error