1501
|
Rausch S, Kruck S, Stenzl A, Bedke J. IMA901 for metastatic renal cell carcinoma in the context of new approaches to immunotherapy. Future Oncol 2014; 10:937-48. [DOI: 10.2217/fon.14.61] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
ABSTRACT: The promising option of immunotherapy for metastatic renal cell carcinoma has evolved from rather unspecific approaches to a specific activation of an anti-tumor T-cell response. The latest step is a synthetic peptide vaccine called IMA901, which demonstrated a clear association between a provoked T-cell response and a prolonged overall survival. The results of IMA901 for the treatment of metastatic renal cell carcinoma are discussed together with new approaches to immunotherapy, such as local and systemic immunomodulation with adjuvants, checkpoint inhibitors, classical chemotherapeutics, such as cyclophosphamide or tyrosine kinase inhibitors. The capability of theses substances to modulate leukocytes subsets, such as myeloid-derived suppressor cells, Tregs or Th17 cells, are outlined together with the possibility to combine them with tumor vaccination strategies to achieve a higher cancer specificity and immunogenicity.
Collapse
Affiliation(s)
- Steffen Rausch
- Department of Urology, Eberhard Karls University Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany
| | - Stephan Kruck
- Department of Urology, Eberhard Karls University Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany
| | - Arnulf Stenzl
- Department of Urology, Eberhard Karls University Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jens Bedke
- Department of Urology, Eberhard Karls University Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
1502
|
Abstract
Increasing knowledge about the gut microbiota composition together with a resurgence in attention to the impact of the host immune system on tumor development triggered our interest in exploring how the interplay of the microbiota and the immune system represents an emerging area of interest. Determining how the immune system may alter gut microbiota composition, or the converse, and whether these interactions increase or reduce cancer risk may be relevant to generate more effective colon cancer preventive strategies.
Collapse
Affiliation(s)
- Florencia McAllister
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Franck Housseau
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Cynthia L. Sears
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
1503
|
Plotnikoff GA. Three measurable and modifiable enteric microbial biotransformations relevant to cancer prevention and treatment. Glob Adv Health Med 2014; 3:33-43. [PMID: 24891992 PMCID: PMC4030612 DOI: 10.7453/gahmj.2014.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Interdisciplinary scientific evaluation of the human microbiota has identified three enteric microbial biotransformations of particular relevance for human health and well-being, especially cancer. Two biotransformations are counterproductive; one is productive. First, selective bacteria can reverse beneficial hepatic hydroxylation to produce toxic secondary bile acids, especially deoxycholic acid. Second, numerous bacterial species can reverse hepatic detoxification-in a sense, retoxify hormones and xeonobiotics-by deglucuronidation. Third, numerous enteric bacteria can effect a very positive biotransformation through the production of butyrate, a small chain fatty acid with anti-cancer activity. Each biotransformation is addressed in sequence for its relevance in representative gastrointestinal and extra-intestinal cancers. This is not a complete review of their connection with every type of cancer. The intent is to introduce the reader to clinically relevant microbial biochemistry plus the emerging evidence that links these to both carcinogenesis and treatment. Included is the evidence base to guide counseling for potentially helpful dietary adjustments.
Collapse
Affiliation(s)
- Gregory A Plotnikoff
- Penny George Institute for Health and Healing, Abbott Northwestern Hospital, Minneapolis, Minnesota, United States
| |
Collapse
|
1504
|
Ursell LK, Haiser HJ, Van Treuren W, Garg N, Reddivari L, Vanamala J, Dorrestein PC, Turnbaugh PJ, Knight R. The intestinal metabolome: an intersection between microbiota and host. Gastroenterology 2014; 146:1470-6. [PMID: 24631493 PMCID: PMC4102302 DOI: 10.1053/j.gastro.2014.03.001] [Citation(s) in RCA: 226] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Recent advances that allow us to collect more data on DNA sequences and metabolites have increased our understanding of connections between the intestinal microbiota and metabolites at a whole-systems level. We can also now better study the effects of specific microbes on specific metabolites. Here, we review how the microbiota determines levels of specific metabolites, how the metabolite profile develops in infants, and prospects for assessing a person's physiological state based on their microbes and/or metabolites. Although data acquisition technologies have improved, the computational challenges in integrating data from multiple levels remain formidable; developments in this area will significantly improve our ability to interpret current and future data sets.
Collapse
Affiliation(s)
- Luke K. Ursell
- Department of Chemistry and Biochemistry, University of Colorado at Boulder, Boulder, CO, USA
| | - Henry J. Haiser
- Faculty of Arts and Sciences (FAS) Center for Systems Biology, Harvard University, Cambridge, MA, USA
| | - Will Van Treuren
- Department of Chemistry and Biochemistry, University of Colorado at Boulder, Boulder, CO, USA
| | - Neha Garg
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Departments of Pharmacology, Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Lavanya Reddivari
- Department of Plant Science, Pennsylvania State University, University Park, PA
| | - Jairam Vanamala
- Department of Food Science, Pennsylvania State University, University Park, PA
| | - Pieter C. Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Departments of Pharmacology, Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Peter J. Turnbaugh
- Faculty of Arts and Sciences (FAS) Center for Systems Biology, Harvard University, Cambridge, MA, USA
| | - Rob Knight
- Department of Chemistry and Biochemistry, University of Colorado at Boulder, Boulder, Colorado; BioFrontiers Institute, University of Colorado at Boulder, Boulder, Colorado; Howard Hughes Medical Institute, Boulder, Colorado.
| |
Collapse
|
1505
|
Abstract
Pancreatic cancer is one of the most lethal cancers worldwide. No effective screening methods exist, and available treatment modalities do not effectively treat the disease. Inflammatory conditions such as pancreatitis represent a well-known risk factor for pancreatic cancer development. Yet only in the past 2 decades has pancreatic cancer been recognized as an inflammation-driven cancer, and the precise mechanisms underlying the pathogenic role of inflammation are beginning to be explored in detail. A substantial amount of preclinical and clinical evidence suggests that bacteria are likely to influence this process by activating immune receptors and perpetuating cancer-associated inflammation. The recent explosion of investigations of the human microbiome have highlighted how perturbations of commensal bacterial populations can promote inflammation and promote disease processes, including carcinogenesis. The elucidation of the interplay between inflammation and microbiome in the context of pancreatic carcinogenesis will provide novel targets for intervention to prevent and treat pancreatic cancer more efficiently. Further studies toward this direction are urgently needed.
Collapse
Affiliation(s)
- Constantinos P. Zambirinis
- S. Arthur Localio Laboratory, Departments of Surgery New York University School of Medicine, New York, NY 10016
| | - Smruti Pushalkar
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010
| | - Deepak Saxena
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010
| | - George Miller
- S. Arthur Localio Laboratory, Departments of Surgery New York University School of Medicine, New York, NY 10016
- S. Arthur Localio Laboratory, Departments of Cell Biology New York University School of Medicine, New York, NY 10016
| |
Collapse
|
1506
|
Perez-Chanona E, Jobin C. From promotion to management: the wide impact of bacteria on cancer and its treatment. Bioessays 2014; 36:658-64. [PMID: 24752750 DOI: 10.1002/bies.201400015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In humans, the intestine is the major reservoir of microbes. Although the intestinal microbial community exists in a state of homeostasis called eubiosis, environmental and genetics factors can lead to microbial perturbation or dysbiosis, a state associated with various pathologies including inflammatory bowel diseases (IBD) and colorectal cancer (CRC). Dysbiotic microbiota is thought to contribute to the initiation and progression of CRC. At the opposite end of the spectrum, two recently published studies in Science reveal that the microbiota is essential for chemotherapeutic drug efficacy, suggesting a beneficial microbial function in cancer management. The dichotomy between the beneficial and detrimental roles of the microbiota during cancer initiation, progression, and treatment emphasize the interwoven relationship between bacteria and cancer. Moreover, these findings suggest that the microbiota could be considered as a therapeutic target, not only at the level of cancer prevention, but also during management, i.e. by enhancing the efficacy of chemotherapeutics.
Collapse
Affiliation(s)
- Ernesto Perez-Chanona
- Department of Medicine and Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL, USA; Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | | |
Collapse
|
1507
|
Chen X, Wakefield LM, Oppenheim JJ. Synergistic antitumor effects of a TGFβ inhibitor and cyclophosphamide. Oncoimmunology 2014; 3:e28247. [PMID: 25050195 PMCID: PMC4063140 DOI: 10.4161/onci.28247] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 02/15/2014] [Indexed: 11/19/2022] Open
Abstract
In a mouse model of breast carcinoma, the combination of cyclophosphamide and transforming growth factor β1,2,3 (TGFβ1,2,3)-targeting antibody achieved superior antineoplastic effects. This novel paradigm of synergistic chemoimmunotherapy promises to improve the clinical outcome of cancer patients with micrometastases, and thus deserves further investigation.
Collapse
Affiliation(s)
- Xin Chen
- Basic Science Program; Leidos Biomedical Research, Inc.; Frederick National Laboratory for Cancer Research; Frederick, MD USA ; Laboratory of Molecular Immunoregulation; Cancer Inflammation Program; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Lalage M Wakefield
- Laboratory of Cancer Biology and Genetics; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Joost J Oppenheim
- Laboratory of Molecular Immunoregulation; Cancer Inflammation Program; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| |
Collapse
|
1508
|
Romano E, Michielin O, Voelter V, Laurent J, Bichat H, Stravodimou A, Romero P, Speiser DE, Triebel F, Leyvraz S, Harari A. MART-1 peptide vaccination plus IMP321 (LAG-3Ig fusion protein) in patients receiving autologous PBMCs after lymphodepletion: results of a Phase I trial. J Transl Med 2014; 12:97. [PMID: 24726012 PMCID: PMC4021605 DOI: 10.1186/1479-5876-12-97] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 03/31/2014] [Indexed: 12/03/2022] Open
Abstract
Background Immunotherapy offers a promising novel approach for the treatment of cancer and both adoptive T-cell transfer and immune modulation lead to regression of advanced melanoma. However, the potential synergy between these two strategies remains unclear. Methods We investigated in 12 patients with advanced stage IV melanoma the effect of multiple MART-1 analog peptide vaccinations with (n = 6) or without (n = 6) IMP321 (LAG-3Ig fusion protein) as an adjuvant in combination with lymphodepleting chemotherapy and adoptive transfer of autologous PBMCs at day (D) 0 (Trial registration No: NCT00324623). All patients were selected on the basis of ex vivo detectable MART-1-specific CD8 T-cell responses and immunized at D0, 8, 15, 22, 28, 52, and 74 post-reinfusion. Results After immunization, a significant expansion of MART-1-specific CD8 T cells was measured in 83% (n = 5/6) and 17% (n = 1/6) of patients from the IMP321 and control groups, respectively (P < 0.02). Compared to the control group, the mean fold increase of MART-1-specific CD8 T cells in the IMP321 group was respectively >2-, >4- and >6-fold higher at D15, D30 and D60 (P < 0.02). Long-lasting MART-1-specific CD8 T-cell responses were significantly associated with IMP321 (P < 0.02). At the peak of the response, MART-1-specific CD8 T cells contained higher proportions of effector (CCR7− CD45RA+/−) cells in the IMP321 group (P < 0.02) and showed no sign of exhaustion (i.e. were mostly PD1−CD160−TIM3−LAG3−2B4+/−). Moreover, IMP321 was associated with a significantly reduced expansion of regulatory T cells (P < 0.04); consistently, we observed a negative correlation between the relative expansion of MART-1-specific CD8 T cells and of regulatory T cells. Finally, although there were no confirmed responses as per RECIST criteria, a transient, 30-day partial response was observed in a patient from the IMP321 group. Conclusions Vaccination with IMP321 as an adjuvant in combination with lymphodepleting chemotherapy and adoptive transfer of autologous PBMCs induced more robust and durable cellular antitumor immune responses, supporting further development of IMP321 as an adjuvant for future immunotherapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Serge Leyvraz
- Department of Oncology, Service of Medical Oncology, CHUV BH-06 1011 Lausanne, Switzerland.
| | | |
Collapse
|
1509
|
Poutahidis T, Kleinewietfeld M, Erdman SE. Gut microbiota and the paradox of cancer immunotherapy. Front Immunol 2014; 5:157. [PMID: 24778636 PMCID: PMC3985000 DOI: 10.3389/fimmu.2014.00157] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 03/25/2014] [Indexed: 12/13/2022] Open
Abstract
It is recently shown that beneficial environmental microbes stimulate integrated immune and neuroendocrine factors throughout the body, consequently modulating regulatory T-lymphocyte phenotypes, maintaining systemic immune balance, and determining the fate of preneoplastic lesions toward regression while sustaining whole body good health. Stimulated by a gut microbiota-centric systemic homeostasis hypothesis, we set out to explore the influence of the gut microbiome to explain the paradoxical roles of regulatory T-lymphocytes in cancer development and growth. This paradigm shift places cancer prevention and treatment into a new broader context of holobiont engineering to cultivate a tumor-suppressive macroenvironment.
Collapse
Affiliation(s)
- Theofilos Poutahidis
- Division of Comparative Medicine, Massachusetts Institute of Technology , Cambridge, MA , USA ; Laboratory of Pathology, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki , Thessaloniki , Greece
| | - Markus Kleinewietfeld
- Departments of Neurology and Immunobiology, Yale School of Medicine , New Haven, CT , USA ; Broad Institute, Massachusetts Institute of Technology and Harvard University , Cambridge, MA , USA ; Faculty of Medicine, Dresden University of Technology (TUD) , Dresden , Germany
| | - Susan E Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology , Cambridge, MA , USA
| |
Collapse
|
1510
|
|
1511
|
Vande Voorde J, Balzarini J, Liekens S. Mycoplasmas and cancer: focus on nucleoside metabolism. EXCLI JOURNAL 2014; 13:300-22. [PMID: 26417262 PMCID: PMC4464442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 02/19/2014] [Indexed: 11/26/2022]
Abstract
The standard of care for patients suffering cancer often includes treatment with nucleoside analogues (NAs). NAs are internalized by cell-specific nucleobase/nucleoside transporters and, after enzymatic activation (often one or more phosphorylation steps), interfere with cellular nucleo(s)(t)ide metabolism and DNA/RNA synthesis. Therefore, their efficacy is highly dependent on the expression and activity of nucleo(s)(t)ide-metabolizing enzymes, and alterations thereof (e.g. by down/upregulated expression or mutations) may change the susceptibility to NA-based therapy and/or confer drug resistance. Apart from host cell factors, several other variables including microbial presence may determine the metabolome (i.e. metabolite concentrations) of human tissues. Studying the diversity of microorganisms that are associated with the human body has already provided new insights in several diseases (e.g. diabetes and inflammatory bowel disease) and the metabolic exchange between tissues and their specific microbiota was found to affect the bioavailability and toxicity of certain anticancer drugs, including NAs. Several studies report a preferential colonization of tumor tissues with some mycoplasma species (mostly Mycoplasma hyorhinis). These prokaryotes are also a common source of cell culture contamination and alter the cytostatic activity of some NAs in vitro due to the expression of nucleoside-catabolizing enzymes. Mycoplasma infection may therefore bias experimental work with NAs, and their presence in the tumor microenvironment could be of significance when optimizing nucleoside-based cancer treatment.
Collapse
Affiliation(s)
- Johan Vande Voorde
- Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, blok x - bus 1030, B-3000 Leuven, Belgium
| | - Jan Balzarini
- Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, blok x - bus 1030, B-3000 Leuven, Belgium
| | - Sandra Liekens
- Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, blok x - bus 1030, B-3000 Leuven, Belgium
| |
Collapse
|
1512
|
Vande Voorde J, Sabuncuoğlu S, Noppen S, Hofer A, Ranjbarian F, Fieuws S, Balzarini J, Liekens S. Nucleoside-catabolizing enzymes in mycoplasma-infected tumor cell cultures compromise the cytostatic activity of the anticancer drug gemcitabine. J Biol Chem 2014; 289:13054-65. [PMID: 24668817 DOI: 10.1074/jbc.m114.558924] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The intracellular metabolism and cytostatic activity of the anticancer drug gemcitabine (2',2'-difluoro-2'-deoxycytidine; dFdC) was severely compromised in Mycoplasma hyorhinis-infected tumor cell cultures. Pronounced deamination of dFdC to its less cytostatic metabolite 2',2'-difluoro-2'-deoxyuridine was observed, both in cell extracts and spent culture medium (i.e. tumor cell-free but mycoplasma-containing) of mycoplasma-infected tumor cells. This indicates that the decreased antiproliferative activity of dFdC in such cells is attributed to a mycoplasma cytidine deaminase causing rapid drug catabolism. Indeed, the cytostatic activity of gemcitabine could be restored by the co-administration of tetrahydrouridine (a potent cytidine deaminase inhibitor). Additionally, mycoplasma-derived pyrimidine nucleoside phosphorylase (PyNP) activity indirectly potentiated deamination of dFdC: the natural pyrimidine nucleosides uridine, 2'-deoxyuridine and thymidine inhibited mycoplasma-associated dFdC deamination but were efficiently catabolized (removed) by mycoplasma PyNP. The markedly lower anabolism and related cytostatic activity of dFdC in mycoplasma-infected tumor cells was therefore also (partially) restored by a specific TP/PyNP inhibitor (TPI), or by exogenous thymidine. Consequently, no effect on the cytostatic activity of dFdC was observed in tumor cell cultures infected with a PyNP-deficient Mycoplasma pneumoniae strain. Because it has been reported that some commensal mycoplasma species (including M. hyorhinis) preferentially colonize tumor tissue in cancer patients, our findings suggest that the presence of mycoplasmas in the tumor microenvironment could be a limiting factor for the anticancer efficiency of dFdC-based chemotherapy. Accordingly, a significantly decreased antitumor effect of dFdC was observed in mice bearing M. hyorhinis-infected murine mammary FM3A tumors compared with uninfected tumors.
Collapse
Affiliation(s)
- Johan Vande Voorde
- From the Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, blok x-bus 1030, B-3000 Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
1513
|
Vacchelli E, Aranda F, Eggermont A, Galon J, Sautès-Fridman C, Cremer I, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Chemotherapy with immunogenic cell death inducers. Oncoimmunology 2014; 3:e27878. [PMID: 24800173 PMCID: PMC4008470 DOI: 10.4161/onci.27878] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 01/15/2014] [Accepted: 01/15/2014] [Indexed: 12/22/2022] Open
Abstract
Accumulating evidence suggests that the clinical efficacy of selected anticancer drugs, including conventional chemotherapeutics as well as targeted anticancer agents, originates (at least in part) from their ability to elicit a novel or reinstate a pre-existing tumor-specific immune response. One of the mechanisms whereby chemotherapy can stimulate the immune system to recognize and destroy malignant cells is commonly known as immunogenic cell death (ICD). Cancer cells succumbing to ICD are de facto converted into an anticancer vaccine and as such elicit an adaptive immune response. Several common chemotherapeutics share the ability of triggering ICD, as demonstrated in vaccination experiments relying on immunocompetent mice and syngeneic cancer cells. A large number of ongoing clinical trials involve such ICD inducers, often (but not always) as they are part of the gold standard therapeutic approach against specific neoplasms. In this Trial Watch, we summarize the latest advances on the use of cyclophosphamide, doxorubicin, epirubicin, oxaliplatin, and mitoxantrone in cancer patients, discussing high-impact studies that have been published during the last 13 months as well as clinical trials that have been initiated in the same period to assess the antineoplastic profile of these immunogenic drugs as off-label therapeutic interventions.
Collapse
Affiliation(s)
- Erika Vacchelli
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Université Paris-Sud/Paris XI; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| | - Fernando Aranda
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Université Paris-Sud/Paris XI; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| | | | - Jérôme Galon
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, UMRS1138; Paris, France ; Laboratory of Integrative Cancer Immunology; Centre de Recherche des Cordeliers; Paris, France
| | - Catherine Sautès-Fridman
- Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, UMRS1138; Paris, France ; Equipe 13; Centre de Recherche des Cordeliers; Paris, France
| | - Isabelle Cremer
- Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, UMRS1138; Paris, France ; Equipe 13; Centre de Recherche des Cordeliers; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy; Villejuif, France ; INSERM, U1015; CICBT507; Villejuif, France
| | - Guido Kroemer
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
1514
|
Haiser HJ, Seim KL, Balskus EP, Turnbaugh PJ. Mechanistic insight into digoxin inactivation by Eggerthella lenta augments our understanding of its pharmacokinetics. Gut Microbes 2014; 5:233-8. [PMID: 24637603 PMCID: PMC4063850 DOI: 10.4161/gmic.27915] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The human gut microbiota plays a key role in pharmacology, yet the mechanisms responsible remain unclear, impeding efforts toward personalized medicine. We recently identified a cytochrome-encoding operon in the common gut Actinobacterium Eggerthella lenta that is transcriptionally activated by the cardiac drug digoxin. These genes represent a predictive microbial biomarker for the inactivation of digoxin. Gnotobiotic mouse experiments revealed that increased protein intake can limit microbial drug inactivation. Here, we present a biochemical rationale for how the proteins encoded by this operon might inactivate digoxin through substrate promiscuity. We discuss digoxin signaling in eukaryotic systems, and consider the possibility that endogenous digoxin-like molecules may have selected for microbial digoxin inactivation. Finally, we highlight the diverse contributions of gut microbes to drug metabolism, present a generalized approach to studying microbe-drug interactions, and argue that mechanistic studies will pave the way for the clinical application of this work.
Collapse
Affiliation(s)
- Henry J Haiser
- Faculty of Arts and Sciences (FAS) Center for Systems Biology; Harvard University; Cambridge, MA USA
| | - Kristen L Seim
- Department of Chemistry and Chemical Biology; Harvard University; Cambridge, MA USA
| | - Emily P Balskus
- Department of Chemistry and Chemical Biology; Harvard University; Cambridge, MA USA
| | - Peter J Turnbaugh
- Faculty of Arts and Sciences (FAS) Center for Systems Biology; Harvard University; Cambridge, MA USA,Correspondence to: Peter J Turnbaugh,
| |
Collapse
|
1515
|
Chappert P. Role of SFB in autoimmune arthritis: an example of regulation of autoreactive T cell sensitivity in the gut. Gut Microbes 2014; 5:259-64. [PMID: 24637598 PMCID: PMC4063855 DOI: 10.4161/gmic.28134] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A key role for segmented filamentous bacteria (SFB) has recently been demonstrated in several mouse models of autoimmune diseases, including autoimmune arthritis and multiple sclerosis. The mechanism governing the activation of systemic autoreactive T cell responses by such commensals in the gut, however, remained elusive. In this addendum, we discuss recent results addressing the local regulation of autoreactive T cell sensitivity by these unique bacteria. We found that the presence of SFB in the gut microbiota was sufficient to promote a local inflammatory microenvironment altering the T cell-intrinsic desensitization process normally occurring in response to chronic self-antigen stimulation. In the absence of this key tolerance checkpoint, sustained chronic T cell proliferation, IFNγ production, and B cell activation eventually led to the development of enhanced pathologies in a Th1-driven T cell-transfer model of autoimmune arthritis.
Collapse
|
1516
|
Chen X, Oppenheim JJ. Th17 cells and Tregs: unlikely allies. J Leukoc Biol 2014; 95:723-731. [PMID: 24563509 DOI: 10.1189/jlb.1213633] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 01/29/2014] [Accepted: 02/02/2014] [Indexed: 01/14/2023] Open
Abstract
Identification of CD4+Foxp3+ Tregs and Th17 modified the historical Th1-Th2 paradigm. Currently, the Th17-Tregs dichotomy provides a dominant conceptual framework for the comprehension of immunity/inflammation and tolerance/immunosuppression in an increasing number of diseases. Targeting proinflammatory Th17 cells or immunosuppressive Tregs has been widely considered as a promising therapeutic strategy in the treatment of major human diseases, including autoimmunity and cancer. The efficacy and safety of such therapy rely on a thorough understanding of immunobiology and interaction of these two subsets of Th cells. In this article, we review recent progress concerning complicated interplay of Th17 cells and Tregs There is compelling evidence that Tregs potently inhibit Th1 and Th2 responses; however, the inhibitory effect of Tregs on Th17 responses is a controversial subject. There is increasing evidence showing that Tregs actually promote the differentiation of Th17 cells in vitro and in vivo and consequently, enhanced the functional consequences of Th17 cells, including the protective effect in host defense, as well as detrimental effect in inflammation and in the support of tumor growth. On the other hand, Th17 cells were also the most potent Th subset in the stimulation and support of expansion and phenotypic stability of Tregs in vivo. These results indicate that these two subsets of Th cells reciprocally stimulate each other. This bidirectional crosstalk is largely dependent on the TNF-TNFR2 pathway. These mutual stimulatory effects should be considered in devising future Th17 cell- and Treg-targeting therapy.
Collapse
Affiliation(s)
- Xin Chen
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA; and .,Laboratory of Molecular Immunoregulation, Cancer Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| | - Joost J Oppenheim
- Laboratory of Molecular Immunoregulation, Cancer Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| |
Collapse
|
1517
|
Viaud S, Daillère R, Yamazaki T, Lepage P, Boneca I, Goldszmid R, Trinchieri G, Zitvogel L. Why should we need the gut microbiota to respond to cancer therapies? Oncoimmunology 2014; 3:e27574. [PMID: 24800167 PMCID: PMC4006853 DOI: 10.4161/onci.27574] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 12/18/2013] [Accepted: 12/18/2013] [Indexed: 01/22/2023] Open
Abstract
Cyclophosphamide, one of the most efficient tumoricidal, antiangiogenic, and immunostimulatory drugs employed to date mediates part of its effects through intestinal bacteria, against which the host becomes immunized during treatment. Our recent work suggests that anti-commensal effector pTH17 and memory TH1 CD4+ T-cell responses are indispensable for optimal anticancer effects as mediated by cyclophosphamide.
Collapse
Affiliation(s)
- Sophie Viaud
- Institut National de la Santé et de la Recherche Médicale; U1015; Gustave Roussy; Villejuif, France ; Université Paris-Sud; Kremlin Bicêtre France
| | - Romain Daillère
- Institut National de la Santé et de la Recherche Médicale; U1015; Gustave Roussy; Villejuif, France ; Université Paris-Sud; Kremlin Bicêtre France
| | - Takahiro Yamazaki
- Institut National de la Santé et de la Recherche Médicale; U1015; Gustave Roussy; Villejuif, France
| | - Patricia Lepage
- Institut National de la Recherche Agronomique, Micalis; UMR1319; Jouy-en-Josas, France ; AgroParisTech; Micalis; UMR1319; Jouy-en-Josas, France
| | - Ivo Boneca
- Unit of Biology and Genetics of the Bacterial Cell Wall; Institut Pasteur; Paris, France ; Institut National de la Santé et de la Recherche Médicale; Group Avenir; Paris, France
| | - Romina Goldszmid
- Cancer and Inflammation Program; National Cancer Institute; Frederick, MD USA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program; National Cancer Institute; Frederick, MD USA
| | - Laurence Zitvogel
- Institut National de la Santé et de la Recherche Médicale; U1015; Gustave Roussy; Villejuif, France ; Centre d'Investigation Clinique Biothérapie CICBT 507; Gustave Roussy; Villejuif, France ; Université Paris-Sud; Kremlin Bicêtre France
| |
Collapse
|
1518
|
Carmona-Gutierrez D, Kroemer G, Madeo F. One cell, one love: a journal for microbial research. MICROBIAL CELL 2014; 1:1-5. [PMID: 28357205 PMCID: PMC5349160 DOI: 10.15698/mic2014.01.118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | - Guido Kroemer
- INSERM, U848, Villejuif, France ; Metabolomics Platform, Institut Gustave Roussy, Villejuif, France ; Centre de Recherche des Cordeliers, Paris, France ; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France ; Université Paris Descartes, Paris 5, Paris, France
| | - Frank Madeo
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| |
Collapse
|
1519
|
|
1520
|
|
1521
|
Abstract
Evaluation of: Iida N, Dzutsev A, Stewart CA et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 342, 967-970 (2013). Intestinal microbiota is essential for host physiological processes, including the maintenance of epithelial barrier and immune functions. However, paradoxically, the intestinal microbiota can promote various types of experimental carcinogenesis. The paper under evaluation demonstrates that disruption of the microbiota impairs the response of tumors to CpG-oligonucleotide immunotherapy and platinum chemotherapy in a context-dependent manner. Thus, intestinal microbiota may have a great impact on the tumor response to chemotherapy and/or immunotherapy.
Collapse
Affiliation(s)
- Naofumi Mukaida
- Division of Molecular Bioregualtion, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| |
Collapse
|
1522
|
|
1523
|
|