1
|
Stefan VE, Weber DD, Lang R, Kofler B. Overcoming immunosuppression in cancer: how ketogenic diets boost immune checkpoint blockade. Cancer Immunol Immunother 2024; 74:23. [PMID: 39537934 PMCID: PMC11561221 DOI: 10.1007/s00262-024-03867-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
Immune checkpoint blockade (ICB) is now part of the standard of care in the treatment of many forms of cancer, yet it lacks efficacy in some patients, necessitating adjunct therapies to support the anti-tumor immune response. Ketogenic diets (KDs), i.e., high-fat low-carbohydrate diets, have been shown to have antiproliferative and immunomodulatory effects in various preclinical cancer studies. Here, we review current knowledge of the complex interplay of KDs and the anti-tumor immune response in the context of ICB therapy, to update our understanding of diet-induced immunometabolic reprogramming in cancer. Preclinical cancer studies have revealed increased activation of and infiltration by tumor-fighting immune cells, especially CD8+ T cells, but also M1 macrophages and natural killer cells, in response to a KD regimen. In contrast, immune-suppressive cells such as regulatory CD4+ T lymphocytes, M2 macrophages, and myeloid-derived suppressor cells were reported to be decreased or largely unaffected in tumors of KD-fed mice. KDs also showed synergism with ICB therapy in several preclinical tumor studies. The observed effects are ascribed to the ability of KDs to improve immune cell infiltration and induce downregulation of immune-inhibitory processes, thus creating a more immunogenic tumor microenvironment. The studies reviewed herein show that altering the metabolic composition of the tumor microenvironment by a KD can boost the anti-tumor immune response and diminish even immunotherapy-resistant as well as immunologically "cold" tumors. However, the exact underlying mechanisms remain to be elucidated, requiring further studies before KDs can be successfully implemented as an adjunct tumor therapy to improve survival rates for cancer patients.
Collapse
Affiliation(s)
- Victoria E Stefan
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | - Daniela D Weber
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Roland Lang
- Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
2
|
Peila R, Rohan TE. Circulating levels of biomarkers and risk of ductal carcinoma in situ of the breast in the UK Biobank study. Int J Cancer 2024; 154:1191-1203. [PMID: 38013398 DOI: 10.1002/ijc.34795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/04/2023] [Accepted: 10/12/2023] [Indexed: 11/29/2023]
Abstract
Observational studies have shown associations between circulating levels of various biomarkers (eg, total cholesterol [TC], low-density lipoprotein cholesterol [LDL], insulin-like growth factor-1 [IGF-1], C-reactive protein [CRP] and glycated hemoglobin-1c [HbA1c]) and the risk of invasive breast cancer (IBC). Ductal carcinoma in situ of the breast (DCIS) is a nonobligate precursor of IBC and shares several risk factors with it. However, the relationship between these biomarkers and DCIS risk remains unexplored. We studied the association between circulating levels of TC, LDL-C, high-density lipoprotein cholesterol (HDL-C), Lipoprotein (a) (Lp-(a)), IGF-1, CRP and HbA1c, with the risk of DCIS in 156801women aged 40 to 69 years and breast cancer-free at enrolment when blood samples and information on demographic and health-related factors were collected. Incident cases of DCIS were ascertained during the follow-up via linkage to the UK cancer registries Multivariable-adjusted Cox proportional hazards models were used to estimate the hazard ratios (HRs) and 95% confidence intervals (CIs) for the associations of interest. In all, 969 DCIS incident cases were diagnosed during 11.4 years of follow-up. Total cholesterol was inversely associated with the risk of DCIS (HRquintile(Q)5vsQ1 = 0.47, 95% CI: 0.27-0.82, Ptrend = .008). Conversely, LDL-C was positively associated with DCIS risk (HRQ3vsQ1 = 1.43, 95% CI: 1.01-2.04, HRQ4vsQ1 = 1.60, 95% CI: 1.04-2.47, HRQ5vsQ1 = 2.29, 95% CI: 1.36-3.88, Ptrend = .004). In postmenopausal women, CRP had a weak positive association with DCIS risk, while HbA1c showed a nonlinear association with the risk. These results, in conjunction with those from previous studies on IBC, provide support for the association of several biomarkers with the risk of an early stage of breast cancer.
Collapse
Affiliation(s)
- Rita Peila
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
3
|
Gnocchi D, Nikolic D, Paparella RR, Sabbà C, Mazzocca A. Cellular Adaptation Takes Advantage of Atavistic Regression Programs during Carcinogenesis. Cancers (Basel) 2023; 15:3942. [PMID: 37568758 PMCID: PMC10416974 DOI: 10.3390/cancers15153942] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Adaptation of cancer cells to extreme microenvironmental conditions (i.e., hypoxia, high acidity, and reduced nutrient availability) contributes to cancer resilience. Furthermore, neoplastic transformation can be envisioned as an extreme adaptive response to tissue damage or chronic injury. The recent Systemic-Evolutionary Theory of the Origin of Cancer (SETOC) hypothesizes that cancer cells "revert" to "primitive" characteristics either ontogenically (embryo-like) or phylogenetically (single-celled organisms). This regression may confer robustness and maintain the disordered state of the tissue, which is a hallmark of malignancy. Changes in cancer cell metabolism during adaptation may also be the consequence of altered microenvironmental conditions, often resulting in a shift toward lactic acid fermentation. However, the mechanisms underlying the robust adaptive capacity of cancer cells remain largely unknown. In recent years, cancer cells' metabolic flexibility has received increasing attention among researchers. Here, we focus on how changes in the microenvironment can affect cancer cell energy production and drug sensitivity. Indeed, changes in the cellular microenvironment may lead to a "shift" toward "atavistic" biologic features, such as the switch from oxidative phosphorylation (OXPHOS) to lactic acid fermentation, which can also sustain drug resistance. Finally, we point out new integrative metabolism-based pharmacological approaches and potential biomarkers for early detection.
Collapse
Affiliation(s)
| | | | | | | | - Antonio Mazzocca
- Interdisciplinary Department of Medicine, University of Bari School of Medicine, Piazza G. Cesare, 11, 70124 Bari, Italy; (D.G.); (D.N.); (R.R.P.); (C.S.)
| |
Collapse
|
4
|
Krysko DV, Demuynck R, Efimova I, Naessens F, Krysko O, Catanzaro E. In Vitro Veritas: From 2D Cultures to Organ-on-a-Chip Models to Study Immunogenic Cell Death in the Tumor Microenvironment. Cells 2022; 11:3705. [PMID: 36429133 PMCID: PMC9688238 DOI: 10.3390/cells11223705] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Immunogenic cell death (ICD) is a functionally unique form of cell death that promotes a T-cell-dependent anti-tumor immune response specific to antigens originating from dying cancer cells. Many anticancer agents and strategies induce ICD, but despite their robust effects in vitro and in vivo on mice, translation into the clinic remains challenging. A major hindrance in antitumor research is the poor predictive ability of classic 2D in vitro models, which do not consider tumor biological complexity, such as the contribution of the tumor microenvironment (TME), which plays a crucial role in immunosuppression and cancer evasion. In this review, we describe different tumor models, from 2D cultures to organ-on-a-chip technology, as well as spheroids and perfusion bioreactors, all of which mimic the different degrees of the TME complexity. Next, we discuss how 3D cell cultures can be applied to study ICD and how to increase the translational potential of the ICD inducers. Finally, novel research directions are provided regarding ICD in the 3D cellular context which may lead to novel immunotherapies for cancer.
Collapse
Affiliation(s)
- Dmitri V. Krysko
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Robin Demuynck
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Iuliia Efimova
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Faye Naessens
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Olga Krysko
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Elena Catanzaro
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| |
Collapse
|
5
|
Kareva I, Brown JS. Estrogen as an Essential Resource and the Coexistence of ER+ and ER– Cancer Cells. Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.673082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Diagnosis of estrogen sensitivity in breast cancer is largely predicated on the ratio of ER+ and ER– cancer cells obtained from biopsies. Estrogen is a growth factor necessary for cell survival and division. It can also be thought of as an essential resource that can act in association with other nutrients, glucose, glutamine, fatty acids, amino acids, etc. All of these nutrients, collectively or individually, may limit the growth of the cancer cells (Liebig’s Law of the Minimum). Here we model estrogen susceptibility in breast cancer as a consumer-resource interaction: ER+ cells require both estrogen and glucose as essential resources, whereas ER– only require the general resource. The model predicts that when estrogen is the limiting factor, other nutrients may go unconsumed and available at higher levels, thus permitting the invasion of ER– cells. Conversely, when ER– cells are less efficient on glucose than ER+ cells, then ER– cells limited by glucose may be susceptible to invasion by ER+ cells, provided that sufficient levels of estrogen are available. ER+ cells will outcompete ER– cells when estrogen is abundant, resulting in low concentrations of interstitial glucose within the tumor. In the absence of estrogen, ER– cells will outcompete ER+ cells, leaving a higher concentration of interstitial glucose. At intermediate delivery rates of estrogen and glucose, ER+ and ER– cells are predicted to coexist. In modeling the dynamics of cells in the same tumor with different resource requirements, we can apply concepts and terms familiar to many ecologists. These include: resource supply points, R∗, ZNGI (zero net growth isoclines), resource depletion, and resource uptake rates. Based on the circumstances favoring ER+ vs. ER– breast cancer, we use the model to explore the consequences of therapeutic regimens that may include hormonal therapies, possible roles of diet in changing cancer cell composition, and potential for evolutionarily informed therapies. More generally, the model invites the viewpoint that cancer’s eco-evolutionary dynamics are a consumer-resource interaction, and that other growth factors such as EGFR or androgens may be best viewed as essential resources within these dynamics.
Collapse
|
6
|
Maldonado R, Talana CA, Song C, Dixon A, Uehara K, Weichhaus M. β-hydroxybutyrate does not alter the effects of glucose deprivation on breast cancer cells. Oncol Lett 2020; 21:65. [PMID: 33281976 PMCID: PMC7709568 DOI: 10.3892/ol.2020.12326] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/29/2020] [Indexed: 12/04/2022] Open
Abstract
Ketogenic diets have the potential to lower glucose availability to cancer cells. However, the effect that the resulting increase in ketone bodies has on cancer cells is not fully understood. The present study explored the effect of β-hydroxybutyrate (BHB) on glucose-deprived MCF-7 and T47D breast cancer cells. Cell proliferation was decreased in response to lower glucose conditions, which could not be rescued consistently by 10 or 25 mM BHB supplementation. In addition, gene expression levels were altered when cells were glucose deprived. Reducing glucose availability of cancer cells to 225 mg/l for 4 days significantly decreased the expression of 113 genes and increased the expression of 100 genes in MCF-7 breast cancer cells, and significantly decreased the expression of 425 genes and increased the expression of 447 genes in T47D breast cancer cells. Pathway enrichment analysis demonstrated that glucose deprivation decreased activity of the Hippo-Yap cell signaling pathway in MCF-7 breast cancer cells, whereas it increased the expression of genes in the NRF2-pathaway and genes regulating ferroptosis in T47D breast cancer cells. Treatment of glucose-deprived cells with 10 or 25 mM BHB significantly changed the expression of 14 genes in MCF-7 breast cancer cells and 40 genes in T47D breast cancer cells. No significant pathway enrichment was detected when glucose-deprived cells were treated with BHB. Both cell lines expressed the enzymes (OXCT1/2, BDH1 and ACAT1/2) responsible for metabolizing BHB to acetyl-CoA, yet expression of these enzymes was not altered by either glucose deprivation or BHB treatment. In the publicly available The Cancer Genome Atlas (TCGA), increased expression of ketone body-catabolizing enzymes was observed in various types of cancer based on mRNA expression z-scores. Increased expression of BDH1 and ACAT1 significantly decreased overall survival of patients with breast cancer in TCGA studies, while decreased OXCT1 expression non-significantly decreased overall survival. In conclusion, neither MCF-7 nor T47D breast cancer cells were affected by BHB during glucose deprivation; however, screening of tumors for activation of ketone body-metabolizing enzymes may be able to identify patients that will benefit from ketogenic diet interventions.
Collapse
Affiliation(s)
- Rylee Maldonado
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA
| | - Chloe Adrienna Talana
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA
| | - Cassaundra Song
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA.,Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marchall University, Huntington, WV 25755, USA
| | - Alyssa Dixon
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA
| | - Kahealani Uehara
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA.,Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Michael Weichhaus
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA
| |
Collapse
|
7
|
Klement RJ, Champ CE, Kämmerer U, Koebrunner PS, Krage K, Schäfer G, Weigel M, Sweeney RA. Impact of a ketogenic diet intervention during radiotherapy on body composition: III-final results of the KETOCOMP study for breast cancer patients. Breast Cancer Res 2020; 22:94. [PMID: 32819413 PMCID: PMC7441712 DOI: 10.1186/s13058-020-01331-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/11/2020] [Indexed: 01/09/2023] Open
Abstract
Background Obesity and low muscle mass are associated with worse outcomes of breast cancer patients. We conducted a controlled trial to study the impact of a ketogenic diet (KD) based on natural foods versus an unspecified standard diet (SD) on body composition in breast cancer patients undergoing radiotherapy. Methods Patients with non-metastasized breast cancer were allocated to either the KD (N = 32) or the SD (N = 31) during radiotherapy. Body composition was measured weekly by bioimpedance analysis. Blood parameters and quality of life were assessed before, during, and at the end of radiotherapy. Results A total of 29 KD and 30 SD patients completed the study. During radiotherapy, mean and median fasting BHB concentrations in the KD group were 0.72 and 0.49 mmol/l (range 0.06–4.9) which was significantly higher than those in the SD group (p < 2.2 × 10−16). There was a very small and insignificant increase in body weight and fat mass in the SD group, as well as a decrease of fat free mass. In contrast, patients in the KD group lost body weight and fat free and skeletal muscle mass quickly after diet onset, which for the most part was related to water losses. The KD did not cause further substantial changes in fat free or skeletal muscle mass, but was associated with a gradual decrease of 0.4 kg body weight and fat mass per week (p < 0.0001). The KD significantly decreased free T3 levels by 0.06 pg/ml/week (p = 6.3 × 10−5). Global quality of life remained stable in the SD group but increased in the KD group from a score of 66.7 to 75.0 (p = 0.20). Conclusions In breast cancer patients undergoing curative radiotherapy, a KD based on natural foods is feasible. After initial water losses, the KD tends to reduce body weight and fat mass while preserving fat free and skeletal muscle mass. Trial registration ClinicalTrials.gov identifier: NCT02516501, registered on August 06, 2015.
Collapse
Affiliation(s)
- Rainer J Klement
- Department of Radiation Oncology, Leopoldina Hospital, Robert-Koch-Straße 10, 97422, Schweinfurt, Germany.
| | - Colin E Champ
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Ulrike Kämmerer
- Department of Obstetrics and Gynaecology, University Hospital of Würzburg, Würzburg, Germany
| | - Petra S Koebrunner
- Department of Radiation Oncology, Leopoldina Hospital, Robert-Koch-Straße 10, 97422, Schweinfurt, Germany
| | - Kelley Krage
- Department of Radiation Oncology, Leopoldina Hospital, Robert-Koch-Straße 10, 97422, Schweinfurt, Germany
| | - Gabriele Schäfer
- Department of Radiation Oncology, Leopoldina Hospital, Robert-Koch-Straße 10, 97422, Schweinfurt, Germany
| | - M Weigel
- Department of Obstetrics and Gynaecology, Leopoldina Hospital, Breast Cancer Centre, Schweinfurt, Germany
| | - Reinhart A Sweeney
- Department of Radiation Oncology, Leopoldina Hospital, Robert-Koch-Straße 10, 97422, Schweinfurt, Germany
| |
Collapse
|