Elsevier

Drug Resistance Updates

Volume 59, December 2021, 100787
Drug Resistance Updates

Hypoxia as a driver of resistance to immunotherapy

https://doi.org/10.1016/j.drup.2021.100787Get rights and content

Abstract

Hypoxia, a hallmark of solid tumors, determines the selection of invasive and aggressive malignant clones displaying resistance to radiotherapy, conventional chemotherapy or targeted therapy. The recent introduction of immunotherapy, based on immune checkpoint inhibitors (ICPIs) and chimeric antigen receptor (CAR) T-cells, has markedly transformed the prognosis in some tumors but also revealed the existence of intrinsic or acquired drug resistance. In the current review we highlight hypoxia as a culprit of immunotherapy failure. Indeed, multiple metabolic cross talks between tumor and stromal cells determine the prevalence of immunosuppressive populations within the hypoxic tumor microenvironment and confer upon tumor cells resistance to ICPIs and CAR T-cells. Notably, hypoxia-triggered angiogenesis causes immunosuppression, adding another piece to the puzzle of hypoxia-induced immunoresistance. If these factors concurrently contribute to the resistance to immunotherapy, they also unveil an unexpected Achille’s heel of hypoxic tumors, providing the basis for innovative combination therapies that may rescue the efficacy of ICPIs and CAR T-cells. Although these treatments reveal both a bright side and a dark side in terms of efficacy and safety in clinical trials, they represent the future solution to enhance the efficacy of immunotherapy against hypoxic and therapy-resistant solid tumors.

Section snippets

Introduction: the impact of hypoxia on tumors and response to therapy

Notwithstanding the compensatory neo-angiogenesis, hypoxic areas are a hallmark of rapidly growing tumors, because of the chaotic architecture of the neo-vessels, and the tendency to undergo vascular collapse under the pressure of growing tumor and stroma (Gacche and Assaraf, 2018; Huijbers et al., 2016; Kleibeuker et al., 2012; Nussenbaum and Herman, 2010). Hypoxic areas are heterogeneously distributed within the tumor bulk, because the continuous alternation between vessel formation and

The imprinting of hypoxia on tumor microenvironment reduces the efficacy of immune checkpoint inhibitors

Hypoxia may impair the efficacy of immunotherapy by acting at multiple levels. A hypoxic environment decreases the ratio between anti-tumor immune cells and immunotolerant or immunosuppressive cells. Furthermore, hypoxia directly increases the expression and activity of ICPs and ICP ligands (ICPLs) on immune-cells and tumor cells. The concurrent presence of immunosuppressive cells, anergic effector cells and immunoevasive cancer cells unequivocally reduces the efficacy of ICPIs.

Mitigating intratumor hypoxia to overcome resistance to immune checkpoint inhibitors: a versatile and open therapeutic field

The pharmacological strategies reducing the deleterious effects of hypoxia worked well in preclinical models to improve the efficacy of chemotherapy, radiotherapy and targeted therapies (Graham and Unger, 2018). Starting from these premises, inhibitors of HIF-1α, agents mitigating the effects of hypoxia, reoxygenation methods may work as immune-sensitizer agents as well. Different strategies have been tested.

Although pharmacological inhibitors of HIF are apparently the easiest category of drugs

The cross talk between hypoxia and angiogenesis: another piece of the puzzle determining the activity of immune checkpoint inhibitors

When the tumors grow, new blood vessels form to provide nutrients and O2. However, the newly formed blood vessels are often structurally and morphologically aberrant, and create a TME with persistent or cycling hypoxia, acidosis and high interstitial fluid pressure (Lugano et al., 2020). These conditions impair the extravasation of immune cells and create an immunosuppressive landscape (Pietrobon and Marincola, 2021), but also offer new therapeutic opportunities to combine anti-angiogenic

Implication of hypoxia-driven changes in the efficacy of CAR T-cells

CAR T-cells represent an effective form of adoptive T-cell therapy (ATC), developed to circumvent the immunotolerance of the T-cell repertoire and the MHC restriction, and to direct specific cytotoxicity to a target molecule on malignant cells. In this approach, T-cells isolated from the patient (or from an allogeneic donor) are genetically modified to express a tailored CAR toward a specific tumor antigen. Then, they are expanded and infused into the patient. The first generation of CAR

Conclusions and future perspectives

Hypoxia is a driver of multiple aggressive features in tumors, inducing metabolic rewiring, apoptosis inhibition, cell migration and increased adaptability to unfavorable conditions. The first consequence of these transformations is the higher resistance of hypoxic tumors to chemotherapy and radiotherapy, as well as to other stressful conditions which usually kill normoxic cells including nutrient deprivation, calcium oscillation, endoplasmic reticulum stress (Akman et al., 2021; Belisario et

Funding

CR unit receives funding from the Italian Association of Cancer Research (AIRC; IG21408). ABSR unit is supported by Foundation for Science and Technology (FCT), Portugal (UID/NEU/04539/2019, UIDB/ 04539/2020 and UIDP/04539/2020). J.D.L.R. unit is funded by Fondo de Investigación Sanitaria—Instituto de Salud Carlos III (FIS—ISCIII, Ministry of Health of Spain; PI18/00591 and PT17/0009/0008) and by the FEDER European Union’s program. J.D.L.R. and C. R. are Vice-Chair and Chair of the COST-Action

Acknowledgements

This article is based upon work from COST Action CA17104 STRATAGEM, supported by COST (European Cooperation in Science and Technology) (www.cost.eu).

References (259)

  • J. Chen et al.

    Regulation of PD-L1: a novel role of pro-survival signalling in cancer

    Ann. Oncol.

    (2016)
  • M. Dal Bo et al.

    New insights into the pharmacological, immunological, and CAR-T-cell approaches in the treatment of hepatocellular carcinoma

    Drug Resist. Updates

    (2020)
  • E.V. Dang et al.

    Control of T(H)17/T(reg) balance by hypoxia-inducible factor 1

    Cell

    (2011)
  • Y. Diesendruck et al.

    Novel immune check point inhibiting antibodies in cancer therapy-Opportunities and challenges

    Drug Resist. Updates

    (2017)
  • J. Dong et al.

    Medicinal chemistry strategies to discover P-glycoprotein inhibitors: an update

    Drug Resist. Updates

    (2020)
  • R. Du et al.

    HIF1α induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion

    Cancer Cell

    (2008)
  • A. Elorza et al.

    HIF2α acts as an mTORC1 activator through the amino acid carrier SLC7A5

    Mol. Cell

    (2012)
  • H.K. Eltzschig et al.

    Central role of Sp1-regulated CD39 in hypoxia/ischemia protection

    Blood

    (2009)
  • N. Erin et al.

    Tumor microenvironment and epithelial mesenchymal transition as targets to overcome tumor multidrug resistance

    Drug Resist. Updat.

    (2020)
  • K. Fischer et al.

    Inhibitory effect of tumor cell-derived lactic acid on human T cells

    Blood

    (2007)
  • R.N. Gacche et al.

    Redundant angiogenic signaling and tumor drug resistance

    Drug Resist. Updates

    (2018)
  • A. Giatromanolaki et al.

    Ectonucleotidase CD73 and CD39 expression in non-small cell lung cancer relates to hypoxia and immunosuppressive pathways

    Life Sci.

    (2020)
  • E. Hays et al.

    YY1 regulates cancer cell immune resistance by modulating PD-L1 expression

    Drug Resist. Updat.

    (2019)
  • E.J. Huijbers et al.

    Role of the tumor stroma in resistance to anti-angiogenic therapy

    Drug Resist. Updates

    (2016)
  • N.A. Hussein et al.

    The role of endolysosomal trafficking in anticancer drug resistance

    Drug Resist. Updates

    (2021)
  • Y. Jiang et al.

    Communication between EMT and PD-L1 signaling: new insights into tumor immune evasion

    Cancer Lett.

    (2020)
  • J.V. Joseph et al.

    Hypoxia enhances migration and invasion in glioblastoma by promoting a mesenchymal shift mediated by the HIF1α – ZEB1 axis

    Cancer Lett.

    (2015)
  • W.G. Kaelin et al.

    Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway

    Mol. Cell

    (2008)
  • R.J. Kathawala et al.

    The modulation of ABC transporter-mediated multidrug resistance in cancer: a review of the past decade

    Drug Resist. Updates

    (2015)
  • A. Kawazoe et al.

    Lenvatinib plus pembrolizumab in patients with advanced gastric cancer in the first-line or second-line setting (EPOC1706): an open-label, single-arm, phase 2 trial

    Lancet Oncol.

    (2020)
  • E.A. Kleibeuker et al.

    Combining angiogenesis inhibition and radiotherapy: a double-edged sword

    Drug Resist. Updates

    (2012)
  • J.N. Kochenderfer et al.

    Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19

    Blood

    (2010)
  • E. Kon et al.

    Immune checkpoint inhibitor combinations: current efforts and important aspects for success

    Drug Resist. Updates

    (2019)
  • V.B. Abdul-Salam et al.

    CLIC4/Arf6 pathway: a new lead in BMPRII inhibition in pulmonary hypertension

    Circ. Res.

    (2019)
  • M. Ai et al.

    Tumor hypoxia drives immune suppression and immunotherapy resistance

    J. Immunother. Cancer

    (2015)
  • M. Akman et al.

    Hypoxia, endoplasmic reticulum stress and chemoresistance: dangerous liaisons

    J. Exp. Clin. Cancer Res.

    (2021)
  • E. Allen et al.

    Combined antiangiogenic and anti-PD-L1 therapy stimulates tumor immunity through HEV formation

    Sci. Transl. Med.

    (2017)
  • F. Antonangeli et al.

    Regulation of PD-L1 expression by NF-κB in cancer

    Front. Immunol.

    (2020)
  • A.B. Apolo et al.

    Phase I study of cabozantinib and nivolumab alone or with ipilimumab for advanced or metastatic urothelial carcinoma and other genitourinary tumors

    J. Clin. Oncol.

    (2020)
  • A.M. Arance et al.

    Lenvatinib (len) plus pembrolizumab (pembro) for patients (pts) with advanced melanoma and confirmed progression on a PD-1 or PD-L1 inhibitor: updated findings of LEAP-004

    J. Clin. Oncol.

    (2021)
  • M.B. Atkins et al.

    Patient-reported outcomes from the phase 3 randomized IMmotion151 trial: atezolizumab + bevacizumab vs sunitinib in treatment-naive metastatic renal cell carcinoma

    Clin. Cancer Res.

    (2020)
  • M. Balsamo et al.

    Hypoxia downregulates the expression of activating receptors involved in NK-cell-mediated target cell killing without affecting ADCC

    Eur. J. Immunol.

    (2013)
  • T.M. Bauer et al.

    The oral HIF-2 α inhibitor MK-6482 in patients with advanced clear cell renal cell carcinoma (RCC): updated follow-up of a phase I/II study

    J. Clin. Oncol.

    (2021)
  • D.C. Belisario et al.

    Hypoxia dictates metabolic rewiring of tumors: implications for chemoresistance

    Cells

    (2020)
  • R. Berahovich et al.

    Hypoxia selectively impairs CAR-T cells in vitro

    Cancers (Basel)

    (2019)
  • A. Bex et al.

    A Phase II, single-arm trial of neoadjuvant axitinib plus avelumab in patients with localized renal cell carcinoma who are at high risk of relapse after nephrectomy (NEOAVAX)

    Future Oncol.

    (2019)
  • E. Boedtkjer

    Na(+),HCO(3)(-) cotransporter NBCn1 accelerates breast carcinogenesis

    Cancer Metastasis Rev.

    (2019)
  • E. Bram et al.

    Mutant Gly482 and Thr482 ABCG2 mediate high-level resistance to lipophilic antifolates

    Cancer Chemother. Pharmacol.

    (2006)
  • M.L. Burr et al.

    CMTM6 maintains the expression of PD-L1 and regulates anti-Tumour immunity

    Nature

    (2017)
  • Q. Cai et al.

    Potential strategies against resistance to CAR T-cell therapy in haematological malignancies

    Ther. Adv. Med. Oncol.

    (2020)
  • Cited by (95)

    View all citing articles on Scopus
    1

    Equal contribution.

    View full text