Hypoxia and Cancer: From Bench to Bedside

A special issue of Cancers (ISSN 2072-6694). This special issue belongs to the section "Tumor Microenvironment".

Deadline for manuscript submissions: closed (28 February 2022) | Viewed by 27990

Special Issue Editors


E-Mail Website
Guest Editor
Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
Interests: pharmacokinetics; natural compounds; anticancer agents; drug–drug interactions; cancer biomarkers
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Co-Guest Editor
Department of Pharmaceutical Sciences, University of South Florida, Tampa, FL, USA
Interests: Acquired Drug Resistance; Tumour-Stromal Interactions; Tumour Hypoxia; Experimental Therapeutics; Cancer Immunotherapy

Special Issue Information

Dear Colleagues,

Hypoxia has been established as a feature of most cancers, playing a key role in cancer progression, angiogenesis, metastasis, and resistance to therapy. Targeting tumor hypoxia as a promising strategy for cancer therapy has therefore generated keen interest in recent years.

Tumor hypoxia is a result of oxygen deprivation to the tissue as rapid tumor growth outstrips its oxygen supply. Research surrounding hypoxia in cancer points to its ability to modulate multiple pathways in tumor survival and metastasis, including its roles in inducing pro-survival gene expression changes, angiogenesis, epithelial-to-mesenchymal transition, invasiveness, and metabolic switches (the Warburg effect), as well as its contribution to the suppression of immune reactivity and DNA repair pathways. Furthermore, it is also recognized that hypoxia contributes to chemoresistance, thereby posing a significant obstacle in cancer therapy. Thus, studying key pathways involved in tumor hypoxia could provide much needed insight into cancer treatment.

In this Special Issue, recent advancements in research on tumor hypoxia will be highlighted. The scope of this Special Issue includes studies ranging from the influence of hypoxia on various hallmarks of cancer, to the use of pharmacological inhibitors to suppress hypoxia-mediated oncogenic pathways in cancer therapy. More importantly, well-controlled clinical investigations are urgently needed to accelerate translational research and fuel the transition of pertinent discoveries in preclinical studies to tangible applications in clinical settings.

Dr. Lingzhi Wang
Dr. Qingyu Zhou
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cancers is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • hypoxia
  • HIF-1
  • tumor microenvironment
  • tumor metastasis
  • Warburg effect
  • cancer therapeutics
  • drug resistance
  • radiotherapy resistance

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review, Other

2 pages, 157 KiB  
Editorial
Hypoxia and Cancer: From Bench to Bedside
by Lingzhi Wang and Qingyu Zhou
Cancers 2023, 15(9), 2478; https://doi.org/10.3390/cancers15092478 - 26 Apr 2023
Viewed by 786
Abstract
This Special Issue of Cancers (two original articles, five reviews), presented by international experts in tumor hypoxia, focuses on the role of hypoxia, or low oxygen levels, in the development and progression of cancer [...] Full article
(This article belongs to the Special Issue Hypoxia and Cancer: From Bench to Bedside)

Research

Jump to: Editorial, Review, Other

16 pages, 3155 KiB  
Article
Post-Hypoxic Cells Promote Metastatic Recurrence after Chemotherapy Treatment in TNBC
by Inês Godet, Mahelet Mamo, Andrea Thurnheer, D. Marc Rosen and Daniele M. Gilkes
Cancers 2021, 13(21), 5509; https://doi.org/10.3390/cancers13215509 - 02 Nov 2021
Cited by 11 | Viewed by 3047
Abstract
Hypoxia occurs in 90% of solid tumors and is associated with treatment failure, relapse, and mortality. HIF-1α signaling promotes resistance to chemotherapy in cancer cell lines and murine models via multiple mechanisms including the enrichment of breast cancer stem cells (BCSCs). In this [...] Read more.
Hypoxia occurs in 90% of solid tumors and is associated with treatment failure, relapse, and mortality. HIF-1α signaling promotes resistance to chemotherapy in cancer cell lines and murine models via multiple mechanisms including the enrichment of breast cancer stem cells (BCSCs). In this work, we utilize a hypoxia fate-mapping system to determine whether triple-negative breast cancer (TNBC) cells that experience hypoxia in the primary tumor are resistant to chemotherapy at sites of metastasis. Using two orthotopic mouse models of TNBC, we demonstrate that cells that experience intratumoral hypoxia and metastasize to the lung and liver have decreased sensitivity to doxorubicin and paclitaxel but not cisplatin or 5-FU. Resistance to therapy leads to metastatic recurrence caused by post-hypoxic cells. We further determined that the post-hypoxic cells that metastasize are enriched in pathways related to cancer stem cell gene expression. Overall, our results show that even when hypoxic cancer cells are reoxygenated in the bloodstream they retain a hypoxia-induced cancer stem cell-like phenotype that persists and promotes resistance and eventually recurrence. Full article
(This article belongs to the Special Issue Hypoxia and Cancer: From Bench to Bedside)
Show Figures

Graphical abstract

18 pages, 1832 KiB  
Article
Impact of Hypoxia on Relative Biological Effectiveness and Oxygen Enhancement Ratio for a 62-MeV Therapeutic Proton Beam
by Chun-Chieh Chan, Fang-Hsin Chen and Ya-Yun Hsiao
Cancers 2021, 13(12), 2997; https://doi.org/10.3390/cancers13122997 - 15 Jun 2021
Cited by 8 | Viewed by 2247
Abstract
This study uses the yields of double-strand breaks (DSBs) to determine the relative biological effectiveness (RBE) of proton beams, using cell survival as a biological endpoint. DSB induction is determined when cells locate at different depths (6 positions) along the track of 62 [...] Read more.
This study uses the yields of double-strand breaks (DSBs) to determine the relative biological effectiveness (RBE) of proton beams, using cell survival as a biological endpoint. DSB induction is determined when cells locate at different depths (6 positions) along the track of 62 MeV proton beams. The DNA damage yields are estimated using Monte Carlo Damage Simulation (MCDS) software. The repair outcomes are estimated using Monte Carlo excision repair (MCER) simulations. The RBE for cell survival at different oxygen concentrations is calculated using the repair-misrepair-fixation (RMF) model. Using 60Co γ-rays (linear energy transfer (LET) = 2.4 keV/μm) as the reference radiation, the RBE for DSB induction and enzymatic DSB under aerobic condition (21% O2) are in the range 1.0–1.5 and 1.0–1.6 along the track depth, respectively. In accord with RBE obtained from experimental data, RMF model-derived RBE values for cell survival are in the range of 1.0–3.0. The oxygen enhancement ratio (OER) for cell survival (10%) decreases from 3.0 to 2.5 as LET increases from 1.1 to 22.6 keV/μm. The RBE values for severe hypoxia (0.1% O2) are in the range of 1.1–4.4 as LET increases, indicating greater contributions of direct effects for protons. Compared with photon therapy, the overall effect of 62 MeV proton beams results in greater cell death and is further intensified under hypoxic conditions. Full article
(This article belongs to the Special Issue Hypoxia and Cancer: From Bench to Bedside)
Show Figures

Figure 1

Review

Jump to: Editorial, Research, Other

26 pages, 2938 KiB  
Review
Significance of Specific Oxidoreductases in the Design of Hypoxia-Activated Prodrugs and Fluorescent Turn off–on Probes for Hypoxia Imaging
by Ewelina Janczy-Cempa, Olga Mazuryk, Agnieszka Kania and Małgorzata Brindell
Cancers 2022, 14(11), 2686; https://doi.org/10.3390/cancers14112686 - 29 May 2022
Cited by 10 | Viewed by 2784
Abstract
Hypoxia is one of the hallmarks of the tumor microenvironment and can be used in the design of targeted therapies. Cellular adaptation to hypoxic stress is regulated by hypoxia-inducible factor 1 (HIF-1). Hypoxia is responsible for the modification of cellular metabolism that can [...] Read more.
Hypoxia is one of the hallmarks of the tumor microenvironment and can be used in the design of targeted therapies. Cellular adaptation to hypoxic stress is regulated by hypoxia-inducible factor 1 (HIF-1). Hypoxia is responsible for the modification of cellular metabolism that can result in the development of more aggressive tumor phenotypes. Reduced oxygen concentration in hypoxic tumor cells leads to an increase in oxidoreductase activity that, in turn, leads to the activation of hypoxia-activated prodrugs (HAPs). The same conditions can convert a non-fluorescent compound into a fluorescent one (fluorescent turn off–on probes), and such probes can be designed to specifically image hypoxic cancer cells. This review focuses on the current knowledge about the expression and activity of oxidoreductases, which are relevant in the activation of HAPs and fluorescent imaging probes. The current clinical status of HAPs, their limitations, and ways to improve their efficacy are briefly discussed. The fluorescence probes triggered by reduction with specific oxidoreductase are briefly presented, with particular emphasis placed on those for which the correlation between the signal and enzyme expression determined with biochemical methods is achievable. Full article
(This article belongs to the Special Issue Hypoxia and Cancer: From Bench to Bedside)
Show Figures

Graphical abstract

25 pages, 3934 KiB  
Review
Vitamin D and Hypoxia: Points of Interplay in Cancer
by Ioanna-Maria Gkotinakou, Ilias Mylonis and Andreas Tsakalof
Cancers 2022, 14(7), 1791; https://doi.org/10.3390/cancers14071791 - 31 Mar 2022
Cited by 3 | Viewed by 3034
Abstract
Vitamin D is a hormone that, through its action, elicits a broad spectrum of physiological responses ranging from classic to nonclassical actions such as bone morphogenesis and immune function. In parallel, many studies describe the antiproliferative, proapoptotic, antiangiogenic effects of calcitriol (the active [...] Read more.
Vitamin D is a hormone that, through its action, elicits a broad spectrum of physiological responses ranging from classic to nonclassical actions such as bone morphogenesis and immune function. In parallel, many studies describe the antiproliferative, proapoptotic, antiangiogenic effects of calcitriol (the active hormonal form) that contribute to its anticancer activity. Additionally, epidemiological data signify the inverse correlation between vitamin D levels and cancer risk. On the contrary, tumors possess several adaptive mechanisms that enable them to evade the anticancer effects of calcitriol. Such maladaptive processes are often a characteristic of the cancer microenvironment, which in solid tumors is frequently hypoxic and elicits the overexpression of Hypoxia-Inducible Factors (HIFs). HIF-mediated signaling not only contributes to cancer cell survival and proliferation but also confers resistance to anticancer agents. Taking into consideration that calcitriol intertwines with signaling events elicited by the hypoxic status cells, this review examines their interplay in cellular signaling to give the opportunity to better understand their relationship in cancer development and their prospect for the treatment of cancer. Full article
(This article belongs to the Special Issue Hypoxia and Cancer: From Bench to Bedside)
Show Figures

Figure 1

29 pages, 1267 KiB  
Review
Targeting HIF-2α in the Tumor Microenvironment: Redefining the Role of HIF-2α for Solid Cancer Therapy
by Leah Davis, Matthias Recktenwald, Evan Hutt, Schuyler Fuller, Madison Briggs, Arnav Goel and Nichole Daringer
Cancers 2022, 14(5), 1259; https://doi.org/10.3390/cancers14051259 - 28 Feb 2022
Cited by 28 | Viewed by 6393
Abstract
Inadequate oxygen supply, or hypoxia, is characteristic of the tumor microenvironment and correlates with poor prognosis and therapeutic resistance. Hypoxia leads to the activation of the hypoxia-inducible factor (HIF) signaling pathway and stabilization of the HIF-α subunit, driving tumor progression. The homologous alpha [...] Read more.
Inadequate oxygen supply, or hypoxia, is characteristic of the tumor microenvironment and correlates with poor prognosis and therapeutic resistance. Hypoxia leads to the activation of the hypoxia-inducible factor (HIF) signaling pathway and stabilization of the HIF-α subunit, driving tumor progression. The homologous alpha subunits, HIF-1α and HIF-2α, are responsible for mediating the transcription of a multitude of critical proteins that control proliferation, angiogenic signaling, metastasis, and other oncogenic factors, both differentially and sequentially regulating the hypoxic response. Post-translational modifications of HIF play a central role in its behavior as a mediator of transcription, as well as the temporal transition from HIF-1α to HIF-2α that occurs in response to chronic hypoxia. While it is evident that HIF-α is highly dynamic, HIF-2α remains vastly under-considered. HIF-2α can intensify the behaviors of the most aggressive tumors by adapting the cell to oxidative stress, thereby promoting metastasis, tissue remodeling, angiogenesis, and upregulating cancer stem cell factors. The structure, function, hypoxic response, spatiotemporal dynamics, and roles in the progression and persistence of cancer of this HIF-2α molecule and its EPAS1 gene are highlighted in this review, alongside a discussion of current therapeutics and future directions. Full article
(This article belongs to the Special Issue Hypoxia and Cancer: From Bench to Bedside)
Show Figures

Figure 1

20 pages, 1881 KiB  
Review
Current Understanding of Hypoxia in Glioblastoma Multiforme and Its Response to Immunotherapy
by Jang Hyun Park and Heung Kyu Lee
Cancers 2022, 14(5), 1176; https://doi.org/10.3390/cancers14051176 - 24 Feb 2022
Cited by 25 | Viewed by 4827
Abstract
Hypoxia is a hallmark of glioblastoma multiforme (GBM), the most aggressive cancer of the central nervous system, and is associated with multiple aspects of tumor pathogenesis. For example, hypoxia induces resistance to conventional cancer therapies and inhibits antitumor immune responses. Thus, targeting hypoxia [...] Read more.
Hypoxia is a hallmark of glioblastoma multiforme (GBM), the most aggressive cancer of the central nervous system, and is associated with multiple aspects of tumor pathogenesis. For example, hypoxia induces resistance to conventional cancer therapies and inhibits antitumor immune responses. Thus, targeting hypoxia is an attractive strategy for GBM therapy. However, traditional studies on hypoxia have largely excluded the immune system. Recently, the critical role of the immune system in the defense against multiple tumors has become apparent, leading to the development of effective immunotherapies targeting numerous cancer types. Critically, however, GBM is classified as a “cold tumor” due to poor immune responses. Thus, to improve GBM responsiveness against immunotherapies, an improved understanding of both immune function in GBM and the role of hypoxia in mediating immune responses within the GBM microenvironment is needed. In this review, we discuss the role of hypoxia in GBM from a clinical, pathological, and immunological perspective. Full article
(This article belongs to the Special Issue Hypoxia and Cancer: From Bench to Bedside)
Show Figures

Figure 1

Other

7 pages, 529 KiB  
Commentary
Cancer Therapeutic Targeting of Hypoxia Induced Carbonic Anhydrase IX: From Bench to Bedside
by Paul C. McDonald, Shawn C. Chafe, Claudiu T. Supuran and Shoukat Dedhar
Cancers 2022, 14(14), 3297; https://doi.org/10.3390/cancers14143297 - 06 Jul 2022
Cited by 47 | Viewed by 3050
Abstract
Carbonic Anhydrase IX (CAIX) is a major metabolic effector of tumor hypoxia and regulates intra- and extracellular pH and acidosis. Significant advances have been made recently in the development of therapeutic targeting of CAIX. These approaches include antibody-based immunotherapy, as well as use [...] Read more.
Carbonic Anhydrase IX (CAIX) is a major metabolic effector of tumor hypoxia and regulates intra- and extracellular pH and acidosis. Significant advances have been made recently in the development of therapeutic targeting of CAIX. These approaches include antibody-based immunotherapy, as well as use of antibodies to deliver toxic and radioactive payloads. In addition, a large number of small molecule inhibitors which inhibit the enzymatic activity of CAIX have been described. In this commentary, we highlight the current status of strategies targeting CAIX in both the pre-clinical and clinical space, and discuss future perspectives that leverage inhibition of CAIX in combination with additional targeted therapies to enable effective, durable approaches for cancer therapy. Full article
(This article belongs to the Special Issue Hypoxia and Cancer: From Bench to Bedside)
Show Figures

Figure 1

Back to TopTop