Factors affecting tumor responders and predictive biomarkers of toxicities in cancer patients treated with immune checkpoint inhibitors

https://doi.org/10.1016/j.intimp.2020.106628Get rights and content

Highlights

  • Factors influencing tumor responders treated with ICIs.

  • Predictive biomarkers of irAEs.

  • New potential mechanisms of resistance to ICI therapy.

Abstract

Cancer immunotherapy has brought a great revolution in the treatment of advanced human cancer. Immune checkpoint inhibitors (ICIs) that target cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and the programmed cell death protein 1 pathway (PD-1/PD-L1) have been widely administrated in the past years and demonstrated promising in a variety of malignancies. While some patients show benefit from ICIs, others do not respond or even develop resistance to these therapies. Among the responders, the treatments are consequently accompanied with immune-related adverse effects (irAEs), which are diverse in their effected organs, degree of severity and timing. Some of the toxicities are fatal and result in discontinuance of immunotherapy. The toxicity profile from anti-CTLA-4 to anti-PD-1/PD-L1 immunotherapies is distinct from those caused by conventional anticancer therapies, though their presentation may be similar. In order to better help clinicians recognize, monitor and manage irAEs in a growing population of cancer patients who are receiving ICI therapy, this article summarizes the FDA approved ICIs and focuses on (1) existing toxic evidence related to ICIs, (2) occurrence of irAEs, (3) factors influencing tumor responders treated with ICIs, (4) predictive biomarkers of irAEs, and (5) new potential mechanisms of resistance to ICI therapy.

Introduction

Cancer patients at their early disease stage are more likely to well respond to most of conventional regimens with a favorable prognosis, whereas those with an advanced disease do not. Immunotherapy is a novel approach developed in the last decade and approved by the United States Food and Drug Administration (FDA) to treat advanced malignancies, which eradicates cancer cells by reinvigorating immune system, activating tumor antigen-specific cytotoxic T lymphocytes (CTLs) with improved clinical outcome in cancer.

Emergence of immune checkpoint blockades opens a new era of cancer therapy. 2018 Nobel Prize in Physiology or Medicine in the discovery of cancer therapy by inhibition of negative immune regulation further ignites enthusiasm to immunotherapeutic for more cancer types [1]. Monoclonal antibodies (mAbs) targeting CTLA-4, PD-1, or PD-L1 have received considerable and broad interest because of their ability to exert endurable clinical response in many first- and second-line regimen-refractory cancers [2], [3], [4]. Some patients generated abiding responses and therefore their survival was improved [5]. Anti-PD1/PD-L1 mAbs increase overall survival (OS) compared to the standard of care in different tumor types. These immune checkpoint inhibitors (ICIs) truly revolutionize the treatment of cancer patients particularly with an advanced disease [6], [7]. Nonetheless, the response rates still remain relatively low [4], [8]. Some patients initially respond to ICI therapy, but develop resistance after relapse [9], [10], [11]. A certain proportion of patients still have a progressive disease in spite of initial response [12], and others do not respond at all [9]. The proportion of patients in the United States who are eligible for these ICIs is approximately 44%, while around 13% respond to these ICIs [4].

Presently, ICIs in combination are being extensively evaluated for potential clinical benefit in a large number of tumor histology. Due to positive outcomes in preliminary trials, nivolumab plus ipilimumab is one of the most enthusiastically investigated combined immunotherapy regimens. Among many cancer types, melanoma is the one with a high response rate to the ICI therapy [13], [14]. On Oct. 22, 2018, Bristol-Myers Squibb (BMS) Company announced that the four-year Phase III CheckMate-067 clinical trial with three arms showed more endurable, long-term survival benefits with the combination of Nivolumab and Ipilimumab versus either Nivolumab or Ipilimumab alone in patients with advanced melanoma [15], [16]. With a minimum follow-up of 48 months, the four-year OS rates were 53% for the Nivolumab plus Ipilimumab in combination, 46% for Nivolumab alone, and 30% for Ipilimumab alone. Additionally, the percentage of the patients experiencing a complete response (CR) was 21% for Nivolumab plus Ipilimumab, 18% for Nivolumab alone, and 5% for Ipilimumab alone [16]. This result suggests that inhibiting both CTLA-4 and PD-1/PD-L1 could synergistically reactivate T cells and were beneficial to patients [17]. Consequently, the combination of the two regimens enhances the efficacy. Indeed, the more recently published results revealed that the combination of Ipilimumab and Nivolumab had a significantly better Progression Free Survival (PFS) compared to chemotherapy in NSCLC [18], with a 42.6% PFS rate for the immunotherapy combo compared to a 13.2% for chemo. Accumulating clinical trial data validate the benefit and rationale of this combinatorial immunotherapy as a powerful new treatment modality for the treatment of cancer.

The most successful immunotherapy to date is the blockade of the immune checkpoints PD-1 and CTLA-4 [19]. However, immune checkpoint blockade can lead to immune-related adverse events (irAEs) that affect any organ system with various clinical presentations. Most of the irAEs are mild, but some are severe and could result in the withdraw of immunotherapy, morbidities or even death. A meta-analysis of 112 clinical trials involving 19,217 patients who received ICIs showed the toxicity-related fatality rates of 0.36% (anti-PD-1), 0.38% (anti-PD-L1), 1.08% (anti-CTLA-4), and 1.23% (PD-1/PD-L1 plus CTLA-4), respectively [20]. There's been a lot of fanfare about the efficacy of the ICIs, but less attention has been paid to the occurrence, prevention, prediction of irAEs, and the resistance mechanisms. Early recognition and treatment of these irAEs represent an important clinical challenge for Oncology physicians. Clinicians across disciplines should not be unaware of these uncommon lethal complications. This article presents the existing toxic evidence related to ICIs, factors influencing tumor responders, prediction and treatment of these irAEs, as well as the new mechanisms of resistance to ICI therapy.

Section snippets

T cell exhaustion and T cell anergy

The immune system consists of innate and adaptive immunity. The innate immune cells target invaders at the first sign of infections or inflammation. Adaptive immune cells, including T cells, recognize and attack specific antigens. Immune evasion is a strategy used by cancer cells to escape host's immune attack, which involves a number of mechanisms, including such as T-cell exhaustion, immunosuppressive cytokines that “cool down” the immune system. T cell exhaustion is a state of T cell

Mechanism of CTLA 4 and PD-1/PD-L1 inhibition

The immune system produces an appropriate immune response against pathogens and cancer. T cell activation plays a pivotal role in killing pathogens and tumor cells. We have reported that T cell activation score, which summarizes weighted average of exhaustion- and activated-related gene expression, is positively associated with patient survival in breast cancer [26]. T cell exhaustion, which is characterized by progressive loss of T cell function, leads to poor prognosis. Recent evidence

Immune checkpoints on activation of anti-tumor immune response

Both the CTLA-4 and PD-1 can participate in T cell dysfunction, but they do not have exactly the same impact on immune system homeostasis, as demonstrated in murine models [34]. CTLA-4 and PD-1 act at different times in the cancer-immunity cycle. CTLA-4 limits T-cell responses early in immune response, primarily in lymphoid tissue; PD-1 limits T-cell response later in immune response, primarily in peripheral tissues. CTLA-4 expressed by T cells and PD-1 expressed by both T cell and other immune

Mechanisms of the ICIs against cancer

Immune checkpoints are actually a normal part of the immune system with the role of preventing immune response from being too strong to destroy healthy cells in the body. Certain protein receptors located on the surface of T cells help distinguish healthy cells from cancer cells. Current ICIs target CTLA-4 and PD-1 receptors. The PD-1 and PD-L1 is a receptor-ligand system and in tumor microenvironment, their engagement results in the blockade of anti-tumor immune responses [47]. PD-1 is mainly

ICIs and their adverse effects

To date, the US FDA has approved seven ICIs as shown in Table 1. Ipilimumab, the first one approved by the US FDA in 2011, targets CTLA-4. From then, several ICIs have been granted marketing authorization. The US FDA approved the second ICI Nivolumab in 2014, which targets PD-1. Combination of Ipilimumab and Nivolumab was approved in 2015. Atezolizumab is the third ICI approved by the US FDA in 2016, which target PD-L1. Pembrolizumab is the fourth ICI that target PD-1 with the US FDA approval

Occurrence of ICI-mediated toxicities and treatment

CTLA-4 and PD-1 immune checkpoints are negative regulators of T-cell immune function, and their expression on T cells leads to immune inhibition. Inhibition of these checkpoints results in an increased activation of the immune system. ICIs including the drugs Ipilimumab that targets CTLA-4 acts as a type of “off switch” on T cells, Pembrolizumab, Nivolumab and Cemiplimab against PD-1 that prevents T cells from recognizing and attacking inflamed tissues and cancer cells. Atezolizumab, Avelumab

Response rates and biomarkers for responders to ICI therapy

As a clinician, it is important to know who will and who will not gain benefit from potential ICI therapy. To determine the percentage of cancer patients who could potentially gain benefit from an ICI drug, Alyson Haslam et al. used annual cancer deaths as a stand-in for annual incidence of advanced or metastatic disease. The results showed that the estimated percentage of patients in the US with cancer eligible for ICIs was 26.86% (95% CI, 1.51–1.57%) in 2015 and increased to an estimated

Mechanisms of resistance to ICIs

Mechanisms of resistance to ICI therapy including innate and acquired resistance are not fully understood. However, durable responses suggestive of long-lasting immunologic memory are commonly observed in large subsets of cancer patients treated with ICIs [90]. Failure of ICI therapy can result from the defects in any of the types as follows: (1) insufficient generation and infiltration of anti-tumor T cells, (2) inadequate function of tumor-specific T cells, (3) impaired formation and

Factors influencing tumor responders treated with ICIs

Some clinicopathological factors such as sex, age, tumor size, and stage, etc. are also predictive markers of ICI therapy. In general, male patients have significantly longer PFS and OS than those female patients [104], and cancer patients whose age between 60 and 75 correlated with better ORR to ICIs [105], [106]. Tumor size is an independent factor that correlates with better ORR and OS for the cancer patients treated with ICIs [107]. In addition, there are some biomarkers that are helpful to

Predictive biomarkers of irAEs

Current ICI therapy has significantly improved the outcome of cancer patients, although the efficacy of the ICI therapy still remains limited. In particular, the therapy only works for a small portion of the cancer patients. Importantly, ICI therapy is also associated with substantial toxicities. Therefore, in order to avoid overtreatment of ICIs and minimize the irAEs development, predictive markers of the irAEs occurrence are important and required [127].

Like the factors that influence tumor

Concluding remarks and outlooks

Over the last decade, cancer immunotherapy has emerged as an attractive addition and alternative to conventional treatment strategies. ICI therapy opens an avenue of cancer immunotherapy with a potent clinical efficacy and prolongs survival in responder patients with cancer. Despite these promising long-term responses, the majority of cancer patients failed to respond to ICI-therapy with primary resistance. More importantly, some of cancer patients have to stop ICI therapy due to irAEs.

References (142)

  • B. El Osta et al.

    Not all immune-checkpoint inhibitors are created equal: Meta-analysis and systematic review of immune-related adverse events in cancer trials

    Crit. Rev. Oncol. Hematol.

    (2017)
  • R. Zappasodi et al.

    Emerging concepts for immune checkpoint blockade-based combination therapies

    Cancer Cell

    (2018)
  • P. Sharma et al.

    Primary, adaptive, and acquired resistance to cancer immunotherapy

    Cell

    (2017)
  • J.M. Pitt et al.

    Resistance mechanisms to immune-checkpoint blockade in cancer: tumor-intrinsic and -extrinsic factors

    Immunity

    (2016)
  • S. George et al.

    Loss of PTEN is associated with resistance to anti-PD-1 checkpoint blockade therapy in metastatic uterine leiomyosarcoma

    Immunity

    (2017)
  • Z.S. Guo

    The 2018 Nobel Prize in medicine goes to cancer immunotherapy (editorial for BMC cancer)

    BMC Cancer

    (2018)
  • L.Z. Shi et al.

    Blockade of CTLA-4 and PD-1 enhances adoptive T-cell therapy efficacy in an ICOS-mediated manner

    Cancer Immunol. Res.

    (2019)
  • J.M. Pacheco et al.

    A changing of the guard: immune checkpoint inhibitors with and without chemotherapy as first line treatment for metastatic non-small cell lung cancer

    Front. Oncol.

    (2019)
  • A. Haslam et al.

    Estimation of the percentage of US patients with cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs

    JAMA Netw. Open

    (2019)
  • S.P. Patel et al.

    A phase II basket trial of dual anti-CTLA-4 and anti-PD-1 blockade in rare tumors (DART SWOG 1609) in patients with nonpancreatic neuroendocrine tumors

    Clin. Cancer Res.

    (2020)
  • Y. Xu et al.

    A.M.E.L.C.C.G. written on behalf of, The association of PD-L1 expression with the efficacy of anti-PD-1/PD-L1 immunotherapy and survival of non-small cell lung cancer patients: a meta-analysis of randomized controlled trials

    Transl. Lung Cancer Res.

    (2019)
  • S.L. Topalian et al.

    Neoadjuvant checkpoint blockade for cancer immunotherapy

    Science

    (2020)
  • V. Longo et al.

    Strategies to improve cancer immune checkpoint inhibitors efficacy, other than abscopal effect. A systematic review

    Cancers (Basel)

    (2019)
  • T.N. Gide et al.

    Primary and acquired resistance to immune checkpoint inhibitors in metastatic melanoma

    Clin. Cancer Res.

    (2018)
  • C.M. Fares et al.

    Mechanisms of resistance to immune checkpoint blockade: why does checkpoint inhibitor immunotherapy not work for all patients?

    Am. Soc. Clin. Oncol. Educ. Book

    (2019)
  • M.J. Mooradian et al.

    What to do when anti-PD-1 therapy fails in patients with melanoma

    Oncology (Williston Park)

    (2019)
  • A. Carretero-Gonzalez et al.

    Analysis of response rate with ANTI PD1/PD-L1 monoclonal antibodies in advanced solid tumors: a meta-analysis of randomized clinical trials

    Oncotarget

    (2018)
  • M.A. Postow et al.

    Immune checkpoint blockade in cancer therapy

    J. Clin. Oncol.

    (2015)
  • M.K. Callahan et al.

    Nivolumab plus ipilimumab in patients with advanced melanoma: updated survival, response, and safety data in a phase I dose-escalation study

    J. Clin. Oncol.

    (2018)
  • J.D. Wolchok et al.

    Overall survival with combined nivolumab and ipilimumab in advanced melanoma

    N. Engl. J. Med.

    (2017)
  • A. Rotte

    Combination of CTLA-4 and PD-1 blockers for treatment of cancer

    J. Exp. Clin. Cancer Res.

    (2019)
  • M.D. Hellmann et al.

    Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer

    Cancer Cell

    (2018)
  • P.A. Beavis et al.

    Dual PD-1 and CTLA-4 checkpoint blockade promotes antitumor immune responses through CD4(+)Foxp3(-) cell-mediated modulation of CD103(+) dendritic cells

    Cancer Immunol. Res.

    (2018)
  • D.Y. Wang et al.

    Fatal toxic effects associated with immune checkpoint inhibitors: a systematic review and meta-analysis

    JAMA Oncol.

    (2018)
  • E.J. Wherry

    T cell exhaustion

    Nat. Immunol.

    (2011)
  • R.H. Schwartz

    T cell anergy

    Annu. Rev. Immunol.

    (2003)
  • L.A. Kalekar et al.

    CD4(+) T cell anergy prevents autoimmunity and generates regulatory T cell precursors

    Nat. Immunol.

    (2016)
  • K. Catakovic et al.

    T cell exhaustion: from pathophysiological basics to tumor immunotherapy

    Cell Commun. Signal.

    (2017)
  • L. Lu et al.

    Elevated T cell activation score is associated with improved survival of breast cancer

    Breast Cancer Res. Treat.

    (2017)
  • M.E. Keir et al.

    PD-1 and its ligands in tolerance and immunity

    Annu. Rev. Immunol.

    (2008)
  • J. Zhao et al.

    Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma

    Nat. Med.

    (2019)
  • C.L. Day et al.

    PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression

    Nature

    (2006)
  • Y. Zhao et al.

    Antigen-Presenting Cell-Intrinsic PD-1 Neutralizes PD-L1 in cis to Attenuate PD-1 Signaling in T Cells

    Cell Rep

    (2018)
  • T. Okazaki et al.

    PD-1 and PD-1 ligands: from discovery to clinical application

    Int. Immunol.

    (2007)
  • M.K. Callahan et al.

    At the bedside: CTLA-4- and PD-1-blocking antibodies in cancer immunotherapy

    J. Leukoc. Biol.

    (2013)
  • A.M. Intlekofer et al.

    At the bench: preclinical rationale for CTLA-4 and PD-1 blockade as cancer immunotherapy

    J. Leukoc. Biol.

    (2013)
  • M. Congreve et al.

    Targeting adenosine A2A receptor antagonism for treatment of cancer

    Expert Opin. Drug Discov.

    (2018)
  • G. Stalhammar et al.

    Expression of immune checkpoint receptors Indoleamine 2,3-dioxygenase and T cell Ig and ITIM domain in metastatic versus nonmetastatic choroidal melanoma

    Cancer Med.

    (2019)
  • Y. Lui et al.

    LAG-3: a very singular immune checkpoint

    Nat. Immunol.

    (2018)
  • M.A. Stanczak et al.

    Self-associated molecular patterns mediate cancer immune evasion by engaging Siglecs on T cells

    J. Clin. Invest.

    (2018)
  • Cited by (13)

    • Photothermic therapy with cuttlefish ink-based nanoparticles in combination with anti-OX40 mAb achieve remission of triple-negative breast cancer

      2023, International Immunopharmacology
      Citation Excerpt :

      Efficient immunotherapy should improve the population of functional tumor-infiltrating lymphocytes (TILs), particularly CD8 T lymphocytes. Higher intratumoral populations of TILs have been recognized of good prognostic value during cancer therapy based on immune checkpoint antibodies (ICAs) such as immune checkpoint blockade (ICB) or immunostimulatory (IS) antibodies [7–10]. However, large solid tumors with a high extracellular matrix (ECM) content, and immune cell-deprived poor tumor microenvironment (TME) demonstrate limited response rates to ICAs, with many patients failing to respond to such therapies [11–14].

    • Immunotherapy for early-stage non-small cell lung cancer: A system review

      2023, Journal of Cancer Research and Therapeutics
    View all citing articles on Scopus
    View full text