Elsevier

Seminars in Cancer Biology

Volume 73, August 2021, Pages 356-376
Seminars in Cancer Biology

The interplay of obesity, gut microbiome and diet in the immune check point inhibitors therapy era

https://doi.org/10.1016/j.semcancer.2021.05.008Get rights and content

Abstract

Immunotherapy has recently emerged as a promising treatment option for many patients, revolutionizing the established therapeutic approach against cancer. Immune checkpoints inhibitors (ICIs) have demonstrated clinical activity in a wide spectrum of malignancies; however, only a minority of patients exhibit durable responses. This response heterogeneity may be partly attributed to host related factors, such as body mass index (BMI), diet and gut microbiome, that have recently emerged as strong influences in ICI responsiveness. Obesity not only directly impacts on cancer promotion but also on the immune homeostasis and the elimination, equilibrium, and escape phases of immune-editing. Paradoxically, emerging clinical data indicate that obese patients are benefited from ICI therapy when compared to normal BMI cancer patients. Interestingly, strong evidence supports the role of the microbiome in cancer immunotherapy, with several recent animal, translational/hybrid and clinical studies demonstrating its influence in the response to ICIs across several malignancies. Noteworthy, nutrition, through its well-established links to obesity, microbiome composition and oncogenicity, may contribute towards leveraging its effects in favor of cancer patients alongside with gold standard treatments. The aim of this review is to delineate the associations of ICIs with obesity, host microbiome and nutrition, and to explore how these factors can be effectively leveraged in enhancing the effectiveness of immunotherapy. More specific aims include the determination of how patients with obesity are differentially affected by ICI therapy; how the host microbiome affects response to ICIs; and how the microbiome itself is modulated by obesity and nutrition. In conclusion, immunometabolism, microbiome and nutrition research present the potential to offer unique tools in unleashing ICIs full potential; providing host-derived, actionable, modifiable targets directly associated with therapeutic outcomes that can be efficiently leveraged. Future efforts, provided that they adhere to robustness of methodology, can facilitate transferring these findings, from bench to bedside.

Introduction

Immunotherapy has recently emerged as a promising treatment option for many patients, revolutionizing the established approach in the combat against cancer. The concept of utilizing properties of the immune system against malignancies dates back several decades [1,2]. In the 1890s, Dr. William Coley noticed that cancer patients who suffered from postsurgical infections exhibited better clinical outcomes. Later in the 1950s, Ehrlich proposed that the immune system constantly exerts thorough surveillance on emergent cancer cells that develop several escape mechanisms to survive. These early investigations ignited the field of cancer immunotherapy. Understanding the processes of immune activation, regulation and interplay with malignant cells was necessary before effectively bringing these concepts in clinical practice. T-cell mediated immunity against cancer cells is the central event in cancer immunotherapy, and occurs as an integration of stimulatory, co-stimulatory and inhibitory signals between T-cells and antigen presenting cells (APCs) [2]. Normally, immune checkpoints function as a ‘break’ that regulates inflammatory responses after T-cells are activated. The first immune checkpoint, the Cytotoxic T-lymphocyte associated protein 4 (CTLA-4), was discovered in the 1980’s by Brunet and colleagues, and its role in cancer immune regulation was elucidated by the seminal work of Krumel and Alison [3,4]. Programmed cell death 1 (PD-1), another immune checkpoint receptor of great importance, was discovered in 1992, and after some years, its ligands, PD-L1 and PD-L2 were identified [5]. Many cancers are currently known to take advantage of these immune checkpoints in order to evade immune surveillance. As a result, blockade of the interactions of CTLA-4 as well as PD-1 with their ligands demonstrate potent anti-tumor effects [6].

Monoclonal antibodies inhibiting immune checkpoints (PD-1 and CTLA-4) have demonstrated clinical activity in a wide spectrum of malignances, including melanoma, non-small cell lung cancer (NSCLC), renal cell, bladder and colorectal cancer, head and neck squamous cell carcinoma, Merkel cell carcinoma, and Hodgkin lymphoma. These therapies have collectively revolutionized the practice of medical oncology [1].

However, CTLA-4, PD-1 and PD-L1 inhibitors may lead to non-specific activation of immune pathways [7], enhancing T cell activation and proliferation, and potentially humoral autoimmunity [7]. This can lead in turn, to inflammatory side effects, termed as immune-related adverse events (irAEs), that comprise a major category of ICI complications [8]. IrAEs can affect every organ system. The gastrointestinal tract, skin, endocrine glands, and liver are among the most frequent tissues of irAEs manifestation, with rash, mucositis, diarrhea and colitis being among the most commonly reported. The central nervous system, cardiovascular, musculoskeletal, pulmonary and hematologic systems can also be affected, albeit less frequently [8].

Despite the promising results observed with the use of ICIs in oncology practice, only a minority of patients exhibit durable responses to ICIs with the remaining either failing to respond or eventually progress while on therapy [9]. This heterogeneity of response has been generally attributed to the individualized degrees of immune resistance that can be either tumor intrinsic or host related [10].

In this context, host related parameters, such as body mass index (BMI) and gut microbiome, have recently emerged as strong influences in ICI therapy responsiveness. In the USA, overweight and obesity may cause 14 % of cancer deaths in men and 20 % in women [11]. Obesity not only directly impacts on cancer promotion but also on the immune homeostasis and the elimination, equilibrium, and escape phases of immune-editing [12]. Paradoxically, emerging clinical data have indicated that patients with obesity are benefited from ICI therapy when compared to normal BMI cancer patients [[13], [14], [15], [16], [17]].

It is well established that the gut microbiome in its interaction with the gastrointestinal mucosa, influences local but also systemic immune responses, at the levels of innate and adaptive immunity [18]. Moreover, strong evidence supports the role of the microbiome in cancer therapy, with several recent animal, translational/hybrid and clinical studies demonstrating its influence on the response to ICIs across several cancers [19]. Ongoing clinical trials aim to elucidate how interventions to the microbiome level may be integrated into clinical practice.

The significance of nutrition for cancer prevention and during active treatment and survivorship is well established [20]. Interestingly, nutrition may affect the gut microbiome, which, in turn, exerts potent different effects on immune function [21,22].

The aim of this review is to delineate the associations of ICIs with obesity, host microbiome and nutrition, and to explore how these factors can be effectively leveraged in enhancing the effectiveness of immunotherapy. More specific aims include the determination of how patients with obesity are differentially affected by ICI therapy and potential causes for these observations; how the host microbiome affects response to ICIs; and how the microbiome itself is modulated by obesity and nutrition.

Section snippets

Key molecules and therapeutic targets

The discovery of the two key molecules-immune checkpoints, the CTLA-4 and the Programmed Cell Death receptor systems, has dramatically changed our comprehension of tumor immune resistance and led to the development of multiple novel therapeutics.

The first key molecule, the CTLA-4 is a protein receptor that behaves as an immune checkpoint being expressed on regulatory T-cells (Tregs) [23]. CTLA-4 is a receptor that has been shown to deliver a co-inhibitory signal during early T-cell activation (

What clinical evidence designated the importance of obesity in the ICI efficacy?

Strong evidence from the International Agency for Research on Cancer (IARC) Working Group suggest that cancers of a plethora of anatomic sites, including colorectal, endometrial, ovarian, esophageal and gastric, renal, pancreatic, hepatocellular and postmenopausal breast are directly linked to excess body weight [59]. Several mechanisms have been hypothesized to mediate this association, including chronic low-grade inflammation/compromised immunity; altered adipocytokine secretion;

The human gastro-intestinal ecosystem and its microbiome

The human gut microbiota encompass approximately 1014 bacteria (i.e. 10 times the number of eukaryotic cells in the human body) and millions of associated genes [99]. These bacteria belong to more than 103 different species, representing more than 3 million genes, and corresponding to a biomass weighting approximately 2 kg [100]. The human gut microbiota constitutes an endocrine organ with a multitude of crucial functions in the organism.

This enormous bacterial population colonizes the

Nutrition, microbiome and ICIs

The science of nutrition has been perceived as a potentially key contributor in obtaining, besides prevention, better prognosis and maximal therapeutic response in cancer patients. The central point of counseling for cancer patients has largely been concentrated on cachexia and lack of appetite as a result of advanced disease or therapy side effects, whereas in practice, cancer patients are more likely to have excess weight than a weight deficit [190]. The practice mainstay advices patients to

Conclusion and future perspectives

ICIs have brought about what cancer researchers have fervently been seeking for many years. Some patients with untreatable cancer enjoy lifespans that overpass even the most wishful predictions. Unfortunately, these outcomes are yet to become the norm. Cancer cells, in a complex interplay with their microenvironment, manage to evade the immune system and survive. The ultimate challenge of the immuno-oncology research community is to maximize the efficacy of immunotherapeutics and ICIs in

Financial support

None.

Transparency document

.

Declaration of Competing Interest

The authors report no declarations of interest.

References (231)

  • S.L. Topalian et al.

    Immune checkpoint blockade: a common denominator approach to cancer therapy

    Cancer Cell

    (2015)
  • J.S. Weber et al.

    Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial

    Lancet Oncol.

    (2015)
  • P. Sharma et al.

    Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy

    Cell

    (2017)
  • K.I. Avgerinos et al.

    Obesity and cancer risk: emerging biological mechanisms and perspectives

    Metabolism

    (2019)
  • S. Ostrand-Rosenberg

    Myeloid derived-suppressor cells: their role in cancer and obesity

    Curr. Opin. Immunol.

    (2018)
  • M. Dalamaga et al.

    The role of extracellular and intracellular Nicotinamide phosphoribosyl-transferase in cancer: diagnostic and therapeutic perspectives and challenges

    Metabolism

    (2018)
  • M. Dalamaga

    Nicotinamide phosphoribosyl-transferase/visfatin: a missing link between overweight/obesity and postmenopausal breast cancer? Potential preventive and therapeutic perspectives and challenges

    Med. Hypotheses

    (2012)
  • G.P. Sotiropoulos et al.

    Chemerin as a biomarker at the intersection of inflammation, chemotaxis, coagulation, fibrinolysis and metabolism in resectable non-small cell lung cancer

    Lung Cancer

    (2018)
  • R. Sender et al.

    Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans

    Cell

    (2016)
  • 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)
  • P. Sharma et al.

    The future of immune checkpoint therapy

    Science

    (2015)
  • M.F. Krummel et al.

    CD28 and CTLA-4 have opposing effects on the response of T ceils to stimulation

    J. Exp. Med.

    (1995)
  • J.F. Brunet et al.

    A new member of the immunoglobulin superfamily-CTLA-4

    Nature

    (1988)
  • M. Ramos-Casals et al.

    Immune-related adverse events of checkpoint inhibitors

    Nat. Rev. Dis. Primers

    (2020)
  • M.A. Postow et al.

    Immune-related adverse events associated with immune checkpoint blockade

    N. Engl. J. Med.

    (2018)
  • R.W. Jenkins et al.

    Mechanisms of resistance to immune checkpoint inhibitors

    Br. J. Cancer

    (2018)
  • S.H. Preston et al.

    Obesity paradox: conditioning on disease enhances biases in estimating the mortality risks of obesity

    Epidemiology

    (2014)
  • T.D. Kanneganti et al.

    Immunological complications of obesity

    Nat. Immunol.

    (2012)
  • G. Kichenadasse et al.

    Association between body mass index and overall survival with immune checkpoint inhibitor therapy for advanced non-small cell lung cancer

    JAMA Oncol.

    (2019)
  • A. Cortellini et al.

    A multicenter study of body mass index in cancer patients treated with anti-PD-1/PD-L1 immune checkpoint inhibitors: when overweight becomes favorable

    J. Immunother. Cancer

    (2019)
  • G.S. Naik et al.

    Complex inter-relationship of body mass index, gender and serum creatinine on survival: exploring the obesity paradox in melanoma patients treated with checkpoint inhibition

    J. Immunother. Cancer

    (2019)
  • A.A. Kolodziejczyk et al.

    Diet–microbiota interactions and personalized nutrition

    Nat. Rev. Microbiol.

    (2019)
  • M.G. Rooks et al.

    Gut microbiota, metabolites and host immunity

    Nat. Rev. Immunol.

    (2016)
  • V. Shankaran et al.

    IFNγ, and lymphocytes prevent primary tumour development and shape tumour immunogenicity

    Nature

    (2001)
  • O.S. Qureshi et al.

    Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4

    Science

    (2011)
  • K. Wing et al.

    CTLA-4 control over Foxp3+ regulatory T cell function

    Science

    (2008)
  • C.A. Chambers et al.

    Secondary but not primary T cell responses are enhanced in CTLA-4-deficient CD8+ T cells

    Eur. J. Immunol.

    (1998)
  • M.F. Krummel et al.

    CTLA-4 engagement inhibits IL-2 accumulation and cell cycle progression upon activation of resting T cells

    J. Exp. Med.

    (1996)
  • S.C. Wei et al.

    Distinct cellular mechanisms underlie Anti-CTLA-4 and Anti-PD-1 checkpoint blockade

    Cell

    (2017)
  • Center for Drug Evaluation and Research. Drug Approvals and Databases [Internet]. U.S. Food and Drug Administration....
  • Y. Latchman et al.

    PD-L2 is a second ligand for PD-1 and inhibits T cell activation

    Nat. Immunol.

    (2001)
  • G.J. Freeman et al.

    Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation

    J. Exp. Med.

    (2000)
  • L.M. Francisco et al.

    PD-L1 regulates the development, maintenance, and function of induced regulatory T cells

    J. Exp. Med.

    (2009)
  • C. Blank et al.

    PD-L1/B7H-1 inhibits the effector phase of tumor rejection by t cell receptor (TCR) transgenic CD8+ t cells

    Cancer Res.

    (2004)
  • Y. Iwai et al.

    PD-1 inhibits antiviral immunity at the effector phase in the liver

    J. Exp. Med.

    (2003)
  • H. Dong et al.

    Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion

    Nat. Med.

    (2002)
  • R.K. Vaddepally et al.

    Review of indications of FDA-Approved immune checkpoint inhibitors per NCCN guidelines with the level of evidence

    Cancers (Basel)

    (2020)
  • M.R. Migden et al.

    PD-1 blockade with cemiplimab in advanced cutaneous squamous-cell carcinoma

    N. Engl. J. Med.

    (2018)
  • S. Ladoire et al.

    Combined evaluation of LC3B puncta and HMGB1 expression predicts residual risk of relapse after adjuvant chemotherapy in breast cancer

    Autophagy.

    (2015)
  • J. Fucikova et al.

    Calreticulin expression in human non-small cell lung cancers correlates with increased accumulation of antitumor immune cells and favorable prognosis

    Cancer Res.

    (2016)
  • Cited by (28)

    • Pathogenic mitochondrial dysfunction and metabolic abnormalities

      2021, Biochemical Pharmacology
      Citation Excerpt :

      Although metabolic reprogramming in metastatic renal cell carcinoma elicits a good response to checkpoint immunotherapy [251,274], in cancers such as urinary bladder cancer [256,272], metabolic reprogramming and/or mitochondrial dysfunction related to reduced OXPHOS [275] have been recently associated with a poor response to immune checkpoint inhibition [276]. The potential role of the microbiome in checkpoint inhibitor response is most intriguing [245,277–280]. A recent analysis of aging as a disease target suggests that a slowdown in aging that increases life expectancy by only one year could be worth US$38 trillion to society and by 10 years it could be worth US$367 trillion [281].

    View all citing articles on Scopus
    View full text