Your browser doesn't support javascript.
loading
Improving combination therapies: targeting A2B-adenosine receptor to modulate metabolic tumor microenvironment and immunosuppression.
Evans, Jason V; Suman, Shankar; Goruganthu, Mounika Uttam L; Tchekneva, Elena E; Guan, Shuxiao; Arasada, Rajeswara Rao; Antonucci, Anneliese; Piao, Longzhu; Ilgisonis, Irina; Bobko, Andrey A; Driesschaert, Benoit; Uzhachenko, Roman V; Hoyd, Rebecca; Samouilov, Alexandre; Amann, Joseph; Wu, Ruohan; Wei, Lai; Pallerla, Aaditya; Ryzhov, Sergey V; Feoktistov, Igor; Park, Kyungho P; Kikuchi, Takefumi; Castro, Julio; Ivanova, Alla V; Kanagasabai, Thanigaivelan; Owen, Dwight H; Spakowicz, Daniel J; Zweier, Jay L; Carbone, David P; Novitskiy, Sergey V; Khramtsov, Valery V; Shanker, Anil; Dikov, Mikhail M.
Afiliação
  • Evans JV; Department of Internal Medicine, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
  • Suman S; Department of Pathology, Anatomy, and Laboratory Medicine, School of Medicine, West Virginia University, Morgantown, WV, USA.
  • Goruganthu MUL; Department of Internal Medicine, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
  • Tchekneva EE; Department of Internal Medicine, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
  • Guan S; Department of Internal Medicine, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
  • Arasada RR; Department of Internal Medicine, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
  • Antonucci A; Department of Internal Medicine, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
  • Piao L; Pfizer Inc, New York, NY, USA.
  • Ilgisonis I; Department of Internal Medicine, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
  • Bobko AA; Department of Internal Medicine, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
  • Driesschaert B; N.V. Sklifosovsky Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.
  • Uzhachenko RV; In Vivo Multifunctional Magnetic Resonance Center, West Virginia University, Morgantown, WV, USA.
  • Hoyd R; Department of Biochemistry, West Virginia University, Morgantown, WV, USA.
  • Samouilov A; In Vivo Multifunctional Magnetic Resonance Center, West Virginia University, Morgantown, WV, USA.
  • Amann J; Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA.
  • Wu R; Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA.
  • Wei L; Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
  • Pallerla A; Department of Internal Medicine, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
  • Ryzhov SV; Department of Internal Medicine, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
  • Feoktistov I; Department of Internal Medicine, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
  • Park KP; Department of Internal Medicine, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
  • Kikuchi T; Department of Internal Medicine, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
  • Castro J; Department of Internal Medicine, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
  • Ivanova AV; Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA.
  • Kanagasabai T; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, TN, USA.
  • Owen DH; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, TN, USA.
  • Spakowicz DJ; Division of Gastroenterology, Department of Internal Medicine, Sapporo Shirakabadai Hospital, Sapporo, Japan.
  • Zweier JL; Palobiofarma SL, Barcelona, Spain.
  • Carbone DP; Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA.
  • Novitskiy SV; School of Graduate Studies, Meharry Medical College, Nashville, TN, USA.
  • Khramtsov VV; Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA.
  • Shanker A; School of Graduate Studies, Meharry Medical College, Nashville, TN, USA.
  • Dikov MM; Department of Internal Medicine, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
J Natl Cancer Inst ; 115(11): 1404-1419, 2023 11 08.
Article em En | MEDLINE | ID: mdl-37195421
ABSTRACT

BACKGROUND:

We investigated the role of A2B-adenosine receptor in regulating immunosuppressive metabolic stress in the tumor microenvironment. Novel A2B-adenosine receptor antagonist PBF-1129 was tested for antitumor activity in mice and evaluated for safety and immunologic efficacy in a phase I clinical trial of patients with non-small cell lung cancer.

METHODS:

The antitumor efficacy of A2B-adenosine receptor antagonists and their impact on the metabolic and immune tumor microenvironment were evaluated in lung, melanoma, colon, breast, and epidermal growth factor receptor-inducible transgenic cancer models. Employing electron paramagnetic resonance, we assessed changes in tumor microenvironment metabolic parameters, including pO2, pH, and inorganic phosphate, during tumor growth and evaluated the immunologic effects of PBF-1129, including its pharmacokinetics, safety, and toxicity, in patients with non-small cell lung cancer.

RESULTS:

Levels of metabolic stress correlated with tumor growth, metastasis, and immunosuppression. Tumor interstitial inorganic phosphate emerged as a correlative and cumulative measure of tumor microenvironment stress and immunosuppression. A2B-adenosine receptor inhibition alleviated metabolic stress, downregulated expression of adenosine-generating ectonucleotidases, increased expression of adenosine deaminase, decreased tumor growth and metastasis, increased interferon γ production, and enhanced the efficacy of antitumor therapies following combination regimens in animal models (anti-programmed cell death 1 protein vs anti-programmed cell death 1 protein plus PBF-1129 treatment hazard ratio = 11.74 [95% confidence interval = 3.35 to 41.13], n = 10, P < .001, 2-sided F test). In patients with non-small cell lung cancer, PBF-1129 was well tolerated, with no dose-limiting toxicities; demonstrated pharmacologic efficacy; modulated the adenosine generation system; and improved antitumor immunity.

CONCLUSIONS:

Data identify A2B-adenosine receptor as a valuable therapeutic target to modify metabolic and immune tumor microenvironment to reduce immunosuppression, enhance the efficacy of immunotherapies, and support clinical application of PBF-1129 in combination therapies.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Carcinoma Pulmonar de Células não Pequenas / Neoplasias Pulmonares Limite: Animals / Humans Idioma: En Revista: J Natl Cancer Inst Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Carcinoma Pulmonar de Células não Pequenas / Neoplasias Pulmonares Limite: Animals / Humans Idioma: En Revista: J Natl Cancer Inst Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Estados Unidos