Submit manuscript...
eISSN: 2373-6372

Gastroenterology & Hepatology: Open Access

Review Article Volume 14 Issue 1

Innovative immune checkpoint inhibitors (ICIs) for cancer treatment: an overview

Manal ME Ahmed

Department of Medicine, University of Science Arts and Technology, USA

Correspondence: Department of Pharmacology, Medical Research and Clinical Studies Institute, Egypt

Received: November 28, 2022 | Published: January 6, 2023

Citation: Ahmed MME. Innovative immune checkpoint inhibitors (ICIs) for cancer treatment: an overview. Gastroenterol Hepatol Open Access. 2023;14(1):1-5. DOI: 10.15406/ghoa.2023.14.00531

Download PDF

Abstract

Cancer immunotherapy has significantly increased patients' chances of survival and quality of life as compared to earlier standards of care (such as chemotherapy, radiation, and surgery). From the metastatic stage to the adjuvant and neoadjuvant settings in many cancer types, immunotherapy has now firmly established itself as a novel pillar of cancer care. In this review article, we emphasise how the development of cancer immunotherapy led to findings that are today considered best practises. We also show out the existing drawbacks and restrictions of cancer checkpoint immunotherapy and the ways in which cutting-edge research is attempting to address these issues.

Keywords: cancer, immunotherapy, immune checkpoint inhibitors, tumor microenvironment, immunoscore

Introduction

The treatment of cancer patients has changed significantly thanks to the field of immuno-oncology.1–6 In the late 19th century, William B. Coley, who is now commonly regarded as the father of immunotherapy, made the first attempts to use the immune system's combative capabilities to cure cancer. He saw that some patients with substantial postoperative wound infections—a frequent occurrence when aseptic technique had not yet been optimized—would spontaneously experience regression of their unresected tumours as an orthopaedic surgeon who operated on patients with bone sarcomas. Coley began injecting mixes of live and dead bacteria, including Streptococcus Pyogenes and Serratia marcescens, into more than a thousand patients in 1891 in an effort to cause sepsis and powerful immunological and antitumor responses. His bacterial cocktail earned the name "Coley's toxin" and is credited with being the first known active cancer immunotherapy treatment.7,8 Coley produced long-lasting complete remissions in a variety of cancers, such as testicular carcinoma, lymphoma, and sarcoma. However, oncologists adopted surgery and radiotherapy as substitute conventional therapies early in the 20th century because Coley's toxin's lack of a recognised mechanism of action and the dangers of purposefully infecting cancer patients with pathogenic bacteria.9

Before Coley's toxin's methods of action could be better understood in relation to the major mediators of sepsis, it would be more than fifty years.10–13 These mediators belong to the cytokine family, which also includes chemokines, interleukins, and interferons.14 The rush to incorporate those fresh discoveries into cancer therapy had begun once more.15–17 With this unique strategy, clinicians and researchers had only little success, occasionally establishing clinical remissions in metastatic renal cell carcinoma with high-dose interleukin 2 (il-2)18 and questionable responses in stages iii and iv melanoma with interferon.19,20 These modest successes were frequently offset by serious negative incidents. Only a tiny, carefully chosen minority of cancer patients would benefit from these innovative delivery techniques, such as pegylation, due to the unpredictable and sporadic immunological reactions they elicited in patients.

With a deeper understanding of the method by which innate immune cells destroy cancer cells—immune surveillance—the field of cancer immunotherapy experienced its next revolutionary wave.21 The field of immuno-oncology has recently entered a new era thanks to the discovery of T cell immune checkpoints like ctla-4 22,23 and PD-124-–27 which also resulted in the awarding of the 2018 Nobel prize in physiology or medicine to Drs. Allison and Honjo. The key job of those hardwired signals is to keep the delicate balance between autoimmunity and immune surveillance against invading infections or aberrant cells. The heightened autoimmunity caused by blocking certain T cell surface receptors provides an immune response against tumours but also raises the risk of autoimmune responses.

In this brief overview, we summaries the current guidelines for cancer immunotherapy, with a particular emphasis on immune checkpoint inhibitors (ICIS), their drawbacks and potential hazards, as well as intriguing innovative approaches.

Checkpoint inhibitors overview

Cancer immuno-editing is the process by which different immune system components safeguard the host against the establishment of primary tumours or promote tumour escape, or both, by either sculpting tumour immunogenicity or attenuating antitumor immune responses.28–30 Immune checkpoints, which are immunological-cell surface receptors that regulate either the activation or suppression of immune responses, strictly regulate the process. On the one hand, activating the immune system is what is needed to prevent tumour growth, but it is also what causes autoimmunity. By upregulating immune activation at different stages of the immunological cycle, the discovery and development of monoclonal antibodies against the inhibitory immune checkpoints ctla-431,32 and PD-133,34 have produced remarkable antitumor responses. Immune checkpoint inhibitor treatments are now frequently recommended for a variety of cancer types (Table 1). Additionally, other clinical trials that are still in progress evaluate how additional agonistic or inhibitory checkpoints may impact outcomes connected to tumours (Table 2). The potential of the checkpoints varies. For instance, the clinical activity of the agonistic OX40 antibody is minimal, whereas the CD28 antibody, even at very low dosages, caused significant cytokine syndrome and required the IC hospitalization of the first six healthy volunteers treated.35,36 In light of this, clinical research is still ongoing to determine the best ICI treatment combination to cause the ideal level of immunological activation.

Agent

Melanoma

NSCLC

RCC

SCHNN

Bladder

Merkel cell carcinoma

Hepatocellular carcinoma

Hodgkin lymphoma

CTLA-4 inhibitor

Ipilimumab

All lines of Tx

             

PD-1 inhibitors

Pembrolizumab

All lines of Tx

All lines of Tx

2nd line Tx

 

2nd line Tx

   

After ASCT

Nivolumab

All lines of Tx

2nd line Tx

 

2nd line Tx

   

2nd line Tx

After ASCT

PD-L1 inhibitors

Atezolizumab

 

2nd line Tx

   

2nd line Tx

     

Avelumab

   

2nd line

   

2nd line Tx

   

Durvalumab

 

After CTxRT
in stage III

Tx

line Tx

2nd line Tx

     

Combination CTLA-4 and PD-1 inhibition

Ipilimumab

1st line Tx

 

1st line Tx

         

nivolumab

               

Table 1 Indications for currently approved immune checkpoint inhibitors in advanced-stage cancers
Source: Health Canada’s Drug Product Database (https://www.canada.ca/en/health-canada/services/drugs-health-products/drug- products/drug-product-database.html).
NSCLC, non-small-cell lung cancer; RCC, renal cell carcinoma (clear cell); SCCHN, squamous-cell carcinoma of head and neck; Tx, treatment; ASCT, autologous stem-cell transplantation; CTxRT, chemoradiotherapy

Target

Drug

Company

Clinical phase

Costimulatory or agonist antibodies

   

4-1BB (CD137)

Utomilumab

Pfizer Canada, Kirkland, QC

I

 

Urelumab

Bristol–Myers Squibb, New York, NY, U.S.A.

I/II

 

INBRX-105

Inhibrx, San Diego, CA, U.S.A.

I

ICOS (CD278)

GSK3359609

GlaxoSmithKline, Mississauga, ON

I/II

 

JTX-2011

Jounce Therapeutics, Cambridge, MA, U.S.A.

I/II

GITR (CD357)

TRX 518-001

Leap Therapeutics, Cambridge, MA, U.S.A.

I/II

 

MK-4166

Merck, Kenilworth, NJ, U.S.A.

I

 

BMS-986156

Bristol–Myers Squibb, New York, NY, U.S.A.

I/II

 

INCAGN01876

Incyte Biosciences International, Wilmington, DE,

I/II

   

U.S.A.

 

CD70

ARGX-110 (cusatuzumab)

Argenx, Breda, Netherlands

I/II

CD27

CDX-1127 (varlilumab)

Celldex Therapeutics, Hampton, NJ, U.S.A.

I/II

OX40 (CD134)

PF-0451860

Pfizer Canada, Kirkland, QC

I/II

 

MEDI0562/6469/6383

AstraZeneca Canada, Mississauga, ON

I

 

GSK3174998

GlaxoSmithKline, Mississauga, ON

I

 

BMS-986178

Bristol–Myers Squibb, New York, NY, U.S.A.

I/II

CD40

CP870893

Pfizer Canada, Kirkland, QC

I

 

APX005M

Bristol–Myers Squibb, New York, NY, U.S.A.

I/II

Co-inhibitory or antagonist antibodies

   

VISTA (B7-H5)

CA-170

Curis, Lexington, MA, U.S.A.

I

CCR4 (CD194)

Mogamulizumab

Kyowa Kirin, Tokyo, Japan

I/II

B7-H3 (CD276)

MGD009

Novartis Pharmaceutical, Ottawa, ON

I

 

8H9

Y-mAbs Therapeutics, New York, NY, U.S.A.

I

TIM-3

TSR-022

Tesaro, Waltham, MA, U.S.A.

I

 

MBG453

Novartis Pharmaceutical, Ottawa, ON

I/ll

 

Sym023

Symphogen A/S, Ballerup, Denmark

I

 

MEDI9447 (oleclumab)

AstraZeneca Canada, Mississauga, ON

I

LAG-3 (CD223)

BMS-986016 (relatlimab)

Bristol–Myers Squibb, New York, NY, U.S.A.

I/II

 

IMP321 (eftilagimod alpha)

Prima BioMed, Sydney, Australia

I

 

LAG525

Novartis Pharmaceutical, Ottawa, ON

I/ll

KIR (2DL1–3)

Lirilumab

Bristol–Myers Squibb, New York, NY, U.S.A.

I/ll

IDO-1,2

Indoximod

NewLink Genetics, Ames, IA, U.S.A.

II

 

Epacadostat

Incyte Biosciences International, Wilmington, DE,

II

   

U.S.A.

 

TIGIT

Tislelizumab

BeiGene, Beijing, P.R.C.

I/II/III

 

BMS-986207

Bristol–Myers Squibb, New York, NY, U.S.A.

I/II

 

MTIG7192A

Genentech, San Francisco, CA, U.S.A.

II/III

 

AB154

Arcus Biosciences, Hayward, CA, U.S.A.

I/II

A2aR

Ciforadenant

Corvus Pharmaceuticals, Burlingame, CA, U.S.A.

I

Transforming

M7824

EMD Serono, Rockland, MA, U.S.A.

I/II

growth factor β

Galunisertib

Eli Lilly and Company, Indianapolis, IN, U.S.A.

II

CD47

TTI-621

Trillium Therapeutics, Mississauga, ON

I

CD73

MEDI9447 (oleclumab)

AstraZeneca Canada, Mississauga, ON

I

Other pathways

     

Toll-like receptors

Poly-ICIC*

Ludwig Institute for Cancer Research, New York, NY,

I

   

U.S.A.

 
 

MGN1703 (lefitolimod)

Mologen, Berlin, Germany

I

 

SD-101

Dynavax Technologies Corporation, Emeryville, CA,

I/II

   

U.S.A.

 
 

DSP-0509

Boston Biomedical, Cambridge, MA, U.S.A.

I/II

 

Rintatolimod

Hemispherx Biopharma, Philadelphia, PA, U.S.A.

II

 

CMP-001

Checkmate Pharmaceuticals, Cambridge, MA, U.S.A.

II

Interleukin 2 receptor

NKTR-214

Nektar Therapeutics, San Francisco, CA, U.S.A.

I/II/III

 

RO6874281

Hoffmann–La Roche, Basel, Switzerland

I/II

 

THOR-707

Synthorx, La Jolla, CA, U.S.A.

I/II

Arginase

CB-1158

Incyte Corporation, Wilmington, DE, U.S.A.

I/II

inhibitors

     

Oncolytic

LTX-315

Lytix Biopharma, Oslo, Norway

II

peptides

     

Interleukin 10

AM0010 (pegilodecakin)

Eli Lilly and Company, Indianapolis, IN, U.S.A.

I/II

Table 2 Agonistic and antagonistic immune checkpoint modulators currently under investigation37
*Poly-ICIC, polyinosinic-polycytidylic acid–poly–L-lysine carboxymethylcellulose.

Validation of immunoscore

Patients' chances of survival and overall quality of life have been significantly improved by cancer immunotherapy. However, not all tumours are created equal, and there are currently very few indicators of toxicity and response. Immuno- oncology is still in its relative infancy despite the quick progress achieved in the field, and there are still many problems and obstacles to be solved. With time, it became clear that the traditional methods for evaluating treatment options during the period of chemotherapy and targeted therapies might not apply to the novel immunotherapies.

Irecist, which accounts for the novel patterns of response seen during immunotherapy, including cancer pseudoprogression.38 was developed by modifying the Response Evaluation Criteria in Solid Tumors (RECIST), which was used to evaluate response to treatments. In the same way that TNM staging has been crucial in guiding treatments in the era of chemotherapy, novel tools are required in the era of cancer immunotherapy. The Immunoscore has already been validated as adding important prognostic information to TNM staging in colon cancer.39 Because of T-cells are currently broadly known as the Key mediators of antitumour efficacy with ICI therapy, using the Immunoscore is an attractive tool to assist guide treatment selection in other cancer types as well. However, this option does not exclude the possible use of additional parameters that might provide further insights into the specifics of each case.

Area to work on for improvement in the immune checkpoint inhibitors’effectiveness and safety

Increasing the efficacy of combination medicines that are already well-established in clinical practise is getting harder. Combining ctla-4 and PD-1 inhibitors has produced an exceptional five-year overall survival rate exceeding 50% in metastatic melanoma.40 The same combination has been linked to an intention-to-treat population overall survival rate of more than 60% at 3years in metastatic renal cell carcinoma.41,42 Few unique combinations in the vast field of ongoing early-phase clinical trials have attained a level of efficacy comparable to those new standards of care. Their safety profiles most definitely need to be enhanced.

In the context of melanoma, the approved induction and regimen dose of combination ICIS—ipilimumab 3mg/kg and nivolumab 1mg/kg every 3 weeks—is linked to a 59% rate of grades 3–4 toxicities.43 Ipilimumab 1mg/kg and nivolumab 3mg/kg every three weeks were the alternative dose methods utilised in CheckMate 511, and preliminary data revealed a considerable improvement in toxicity without a loss of efficacy.44 Predictors and cutting-edge methods to reduce those toxicities are urgently required because iraes can occasionally be linked to mortality and serious lifetime morbidity (such as de novo insulin-dependent diabetes, persistent pituitary dysfunction, or immune-related inflammatory arthropathies).

Finding innovative treatments for patients who are both primary non-responders to ICIS and those who develop secondary resistance to such therapies is another area in which there is a critical need.45–51 Very few therapies have been examined beyond ICI failure, and clinicians frequently follow already approved standards of care for each particular cancer. Early observational data imply that ICIS exposure may modify the responsiveness to conventional treatments administered after progression. For instance, after ICI failure, extremely high chemotherapeutic response rates have occasionally been documented.52,53 Those observations may be a byproduct of immunotherapy, which eliminated the initial inhibition that tumour cells or other immune cells had exerted, followed by the cytotoxic chemotherapy-mediated eradication of the tumour cells. On the other hand, first- line exposure to ICIS may have a negative impact on progression-free survival and the adverse event profiles linked to exposure to targeted therapies (such as BRAF inhibition in melanoma).54–60

Conclusion

In conclusion, combination therapies utilizing checkpoint inhibitors rather than additional new checkpoint inhibitors may be the future of cancer immunotherapy. The current wide "shotgun" strategy, which exposes everyone within the approved indications to ICIS, will be replaced by tailored therapies that are specific to the characteristics that make each cancer and host a particular pairing as a result of advancements in those domains.

Acknowledgments

None.

Conflicts of interest

We declare there is no conflict of interest.

Funding

None.

References

  1. Avgerinos KI, Spyrou N, Mantzoros CS, et al. Obesity and cancer risk: Emerging biological mechanisms and perspectives. Metabolism. 2019;92:121–135.
  2. Mattiuzzi C, Lippi G. Current cancer epidemiology. J Epidemiol Global Health. 2019;9(4):217.
  3. Kim R. Cancer immunoediting: from immune surveillance to immune escape. Immunology. 2007;121(1):1–14.
  4. Hernández–Camarero P, López–Ruiz E, Marchal JA, et al. Cancer: a mirrored room between tumor bulk and tumor microenvironment. J Exp Clin Cancer Res. 2021;40(1):217.
  5. Xiao Y, Yu D. Tumor microenvironment as a therapeutic target in cancer. Pharmacol Ther. 2021;221:107753.     
  6. Whiteside T. The tumor microenvironment and its role in promoting tumor growth. Oncogene. 2008;27(45):5904–5912.
  7. McCarthy EF. The toxins of William B. Coley and the treatment of bone and soft–tissue sarcomas. Iowa Orthop J. 2006;26:154–158.
  8. Ahmed MME. CAR–T cell therapy: current advances and future research possibilities. Journal of Scientific Research in Medical and Biological Sciences. 2021;2(2):86–116.
  9. Decker WK, Safdar A. Bioimmunoadjuvants for the treatment of neoplastic and infectious disease: Coley’s legacy revisited. Cytokine Growth Factor Rev. 2009;20:271–281.
  10. Ahmed MME. Future approaches for Brucellosis vaccines and therapies development based on molecular host–pathogen interaction. Gastroenterology & Hepatology:Open Access. 2022;13(4):145–154.
  11. Ahmed MME. MRSA infections: priorities and future approaches for research. Gastroenterology & Hepatology:Open Access. 2022;13(6):200– 208.
  12. Ahmed MME, Eljakee J, Mahran T. Development of Novel Protective Polyvalent Irradiated Pseudomonas aeruginosa Vaccine for Immuno– Compromised Patients. International Journal of Pharmacology, Phytochemistry and Ethnomedicine. 2021;16:1–10.
  13. Ahmed MME, Eljakee J, Mahran T. Developement of anti– P. aeruginosa immunoglobulin Y antibodies as prophylacic therapy for cystic fibrosis patients. International Journal of Scientific Research in Biological Sciences. 2020;7(2):44–50.
  14. Dinarello CA. Historical insights into cytokines. Eur J Immunol. 2007;37(suppl 1):S34–S45.
  15. Lee S, Margolin K. Cytokines in cancer immunotherapy. Cancers (Basel). 2011;3:3856–93.
  16. Ahmed MME, Soliman R, Eljakee J, et al. Preparation of Hybridomas Producing Monoclonal Antibodies against Aflatoxin B1 as a Tool to Control Hepatocellular Carcinoma. International Journal of Pharmacology, Phytochemistry and Ethnomedicine. 2019;13:1–12.
  17. Longo V, Gnoni A, Casadei Gardini A, et al. Immunotherapeutic approaches for hepatocellular carcinoma. Oncotarget. 2017;8(20):33897–33910.
  18. Nghiem P, Bhatia S, Lipson EJ, et al. Durable tumor regression and overall survival in patients with advanced merkel cell carcinoma receiving pembrolizumab as first–line therapy. J Clin Oncol. 2019;37(9):693–702.
  19. Rosenberg SA. Interleukin 2 for patients with renal cancer. Nat Clin Pract Oncol 2007;4:497. [Comment on: TwardowskiP, Figlin RA. What are the indications for sorafenib treatment In patients with renal cell carcinoma? Nat Clin Pract Oncol. 2007;4:456–457; and Stadler WM, Szmulewitz RZ. Sunitinib—a New standard of care for metastatic renal cell carcinoma. Nat Clin Pract Oncol. 2007;4:458–459.
  20. Di Trolio R, Simeone E, Di Lorenzo G, et al. The use of interferon in melanoma patients: a systematic Review. Cytokine Growth Factor Rev. 2015;26:203–212.
  21. Zhu S, Zhang T, Zheng L, et al. Combination strategies to maximize the benefits of cancer immunotherapy. J Hematol Oncol. 2021;14(1):156.
  22. Buchbinder EI, Desai A. CTLA–4 and PD–1 pathways: similarities, differences, and implications of their inhibition. Am J Clin Oncol. 2016;39:98.
  23. Friedline RH, Brown DS, Nguyen H, et al. CD4+ regulatory T cells require CTLA–4 for the maintenance of systemic tolerance. J Exp Med. 2009;206:421–434.
  24. Akinleye A, Rasool Z. Immune checkpoint inhibitors of PD–L1 as cancer therapeutics. J Hematol Oncol. 2019;12:1–13.
  25. Keir ME, Francisco LM, Sharpe AH. PD-1 and its ligands in T-cell Immunity. Curr Opin Immunol. 2007;19:309–314.
  26. Lotfinejad P, Kazemi T, Mokhtarzadeh A, et al. PD–1/PD–L1 axis importance and tumor microenvironment immune cells. Life Sci. 2020;259:118297.
  27. Keir ME, Liang SC, Guleria I, et al. Tissue expression of PD–L1 mediates peripheral T cell tolerance. J Exp Med. 2006;203:883–895.
  28. O’Donnell JS, Teng MWL, Smyth MJ. Cancer immunoediting and resistance to T cell–based immunotherapy. Nat Rev Clin Oncol. 2019;16:151–167.
  29. Ahmed MME. Novel strategies to improve car-t cells in solid tumors: a mini review. Journal of Scientific Research in Medical and Biological Sciences. 2022;3(3):36–47.
  30. Avgerinos KI, Spyrou N, Mantzoros CS, et al. Obesity and cancer risk: Emerging biological mechanisms and perspectives. Metabolism. 2019;92:121–135.
  31. Vargas FA, Furness AJ, Litchfield K, et al. Fc effector function contributes to the activity of human anti–CTLA–4 antibodies. Cancer Cell. 2018;33:649–663.e4.
  32. Hargadon KM, Johnson CE, Williams CJ. Immune checkpoint blockade therapy for cancer: an overview of FDA–approved immune checkpoint inhibitors. Int Immunopharmacol. 2018;62:29–39.
  33. Miyazaki T, Ishikawa E, Matsuda M, et al. Assessment of PD–1 positive cells on initial and secondary resected tumor specimens of newly diagnosed glioblastoma and its implications on patient outcome. J Neuro Oncol. 2017;133:277–285.
  34. Wang Y, Wang H, Yao H, et al. Regulation of PD–L1: Emerging routes for targeting tumor immune evasion. Front Pharmacol. 2018;9:536.
  35. Suntharalingam G, Perry MR, Ward S, et al. Cytokine storm in a phase 1 trial of the anti–CD28 monoclonal antibody TGN1412. N Engl J Med. 2006;355:1018–1028.
  36. Ogura A, Akiyoshi T, Yamamoto N, et al. Pattern of programmed cell death–ligand 1 expression and CD8–positive T–cell infiltration before and after chemoradiotherapy in rectal cancer. Eur J Cancer. 2018;91:11–20.
  37. Esfahani K, Roudaia L, Buhlaiga N, et al. Review of cancer immunotherapy: From the past, to the present, to the future. Current Oncology. 2020;27 (Supp. 2):587–597.
  38. Seymour L, Bogaerts J, Perrone A, et al. On behalf of the recist Working group. Irecist: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017;18:e143–e152.
  39. Pages F, Mlecnik B, Marliot F, et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet. 2018;391:2128–2139.
  40. Larkin J, Chiarion–Sileni V, Gonzalez R, et al. Five–year survival with combined nivolumab and ipilimumab in advanced Melanoma. N Engl J Med. 2019;381:1535–1546.
  41. Motzer RJ, Rini BI, McDermott DF, et al. On behalf of the CheckMate 214 investigators. Nivolumab plus ipilimumab Versus sunitinib in first–line treatment for advanced renal cell carcinoma: extended follow–up of efficacy and safety results from a randomised, controlled, phase 3 trial. Lancet Oncol. 2019;20(10):1370–1385.
  42. Motzer RJ, Tannir NM, McDermott DF, et al. On behalf of the CheckMate 214 investigators. Nivolumab plus ipilimumab Versus sunitinib in advanced renal–cell carcinoma. N Engl J Med. 2018;378:1277–1290.
  43. Wolchok JD, Chiarion–Sileni V, Gonzalez R, et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2017;377:1345–1356; [Erratum In: Neoadjuvant PD–1 blockade in resectable lung cancer; Nivolumab and ipilimumab in advanced melanoma; Overall survival with combined nivolumab and ipilimumab in advanced melanoma; Prolonged survival in stageiii melanoma with ipilimumab adjuvant therapy; Combined nivolumab and ipilimumab or monotherapy in untreated melanoma; Combined nivolumab and ipilimumab or monotherapy in untreated melanoma; Nivolumab and ipilimumab versus Ipilimumab in untreated melanoma; Rapid eradication of a bulky melanoma mass with one dose of immunotherapy; Genetic basis for clinical response to ctla–4 blockade; Genetic basis for clinical response to ctla–4 blockade in melanoma; Nivolumab plus ipilimumab in advanced melanoma; Safety and tumor responses with lambrolizumab (anti–PD–1) in melanoma; Hepatotoxicity with combination of vemurafenib and ipilimumab. N Engl J Med. 2018;379:2185.
  44. Lebbé C, Meyer N, Mortier L, et al. Evaluation of two dosing regimens for nivolumab in combination with ipilimumab in patients with advanced melanoma: results from the phase iiib/iv CheckMate 511 trial. J Clin Oncol. 2019;37:867–875.
  45. Kim N, Kim HS. Targeting checkpoint receptors and molecules for therapeutic modulation of natural killer cells. Front Immunol. 2018;9:2041.
  46. Wang J, Li J, Tang G, Tian Y, Su S, Li Y. Clinical outcomes and influencing factors of PD–1/PD–L1 in hepatocellular carcinoma. Oncol Lett. 2021;21:279.
  47.  Chen J, Wang J, Xu H. Comparison of atezolizumab, durvalumab, pembrolizumab, and nivolumab as first–line treatment in patients with extensive–stage small cell lung cancer: A systematic review and network metaanalysis. Med (Baltimore). 2021;100:e25180.
  48. Peyrottes A, Ouzaid I, Califano G, et al. Neoadjuvant Immunotherapy for muscle–invasive bladder cancer. Medicina (Kaunas). 2021;57:769.
  49. Rizvi NA, Mazières J, Planchard D, et al. Activity and safety of nivolumab, an anti–PD–1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non–small–cell lung cancer (CheckMate 063): a phase 2, single–arm trial. Lancet Oncol. 2015;16:257–265.
  50. Narayan V, Kahlmeyer A, Dahm P, et al. Pembrolizumab monotherapy versus chemotherapy for treatment of advanced urothelial carcinoma with disease progression during or following platinumcontaining chemotherapy. A cochrane rapid review. Cochrane Database Syst Rev. 2018;7:Cd012838.    
  51. Van Den Ende T, Van Den Boorn HG, Hoonhout NM, et al. Priming the tumor immune microenvironment with chemo (radio) therapy: a systematic review across tumor types. Biochim Biophys Acta (BBA) Reviews Cancer. 2020;1874:188386.
  52. Dwary AD, Master S, Patel A, et al. Excellent response to chemotherapy post immunotherapy. Oncotarget. 2017;8(53):91795–802.
  53. Aguilera JV, Paludo J, Bangalore A, et al. Chemoimmunotherapy combination after PD–1 inhibitor failure to improve clinical outcomes in metastatic melanoma patients [abstract 9558]. J Clin Oncol. 2018;36.
  54. Xia CY, Wang DY, Mason R, et al. Activity of targeted therapy after failure of first–line immunotherapy in BRAF–mutant metastatic melanoma [abstract 9532]. J Clin Oncol. 2018;36.
  55. Liu D, Jenkins RW, Sullivan RJ. Mechanisms of resistance to immune checkpoint blockade. Am J Clin Dermatol. 2019;20:41–54.
  56. Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discovery. 2018;8:1069–1086.
  57. Mokhtari RB, Homayouni TS, Baluch N, et al. Combination therapy in combating cancer. Oncotarget. 2017;8:38022.
  58. Zhu S, Zhang T, Zheng L, et al. Combination strategies to maximize the benefits of cancer immunotherapy. J Hematol Oncol. 2021;14:156.
  59. Najafi M, Majidpoor J, Toolee H, et al. The current knowledge concerning solid cancer and therapy. J Biochem Mol Toxicol. 2021;35:e22900.
  60. Liang JL, Luo GF, Chen WH, et al. Recent advances in engineered materials for immunotherapy–involved combination cancer therapy. Adv Mater. 2021;33:e2007630.
Creative Commons Attribution License

©2023 Ahmed. This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.

Citations