1
|
Borazanci EH, Bahary N, Chung V, Huyck TK, Kio EA, Chiorean EG, Skeel RT, Alese OB, Cardin DB, Fountzilas C, Hanna WT, Leal AD, Lee V, Noonan AM, Philip PA, Wainberg ZA, Pashova H, Mann G, Oberstein PE. Relacorilant plus nab-paclitaxel for the treatment of metastatic pancreatic ductal adenocarcinoma: results from the open-label RELIANT study. Oncologist 2024; 29:957-965. [PMID: 39191530 PMCID: PMC11546724 DOI: 10.1093/oncolo/oyae210] [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: 03/29/2024] [Accepted: 06/15/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Modulation of glucocorticoid receptor (GR) activity in tumor cells enhances chemotherapy efficacy. We evaluated the selective GR modulator relacorilant plus nab-paclitaxel in patients with metastatic pancreatic ductal adenocarcinoma (mPDAC) who had received at least 2 prior therapy lines. PATIENTS AND METHODS In this open-label, single-arm, phase III study, patients received once-daily oral relacorilant (100 mg, titrated to 150 mg in 25 mg increments/cycle) and nab-paclitaxel (80 mg/m2) on days 1, 8, and 15 of 28-day cycles. The primary efficacy endpoint was objective response rate (ORR) by blinded independent central review. Progression-free survival (PFS), overall survival (OS), target gene modulation, and safety were also assessed. RESULTS Of 43 patients enrolled, 31 were evaluable for ORR (12 did not reach first postbaseline radiographic assessment). An interim analysis to assess whether ORR was ≥10% showed no confirmed responses and the study was discontinued. Two (6.5%) patients attained unconfirmed partial responses and 15 (48.4%) had stable disease. Fourteen of 31 (45.2%) patients had reductions in target lesion size, despite prior nab-paclitaxel exposure in 12 of the 14. Median PFS and OS were 2.4 months (95% CI, 1.4-4.2) and 3.9 months (95% CI, 2.8-4.9), respectively. The most common adverse events were fatigue and nausea. RNA analysis confirmed that relacorilant plus nab-paclitaxel suppressed 8 cortisol target genes of interest. CONCLUSION Relacorilant plus nab-paclitaxel showed modest antitumor activity in heavily pretreated patients with mPDAC, with no new safety signals. Studies of this combination in other indications with a high unmet medical need are ongoing.
Collapse
Affiliation(s)
| | - Nathan Bahary
- Allegheny Health Network Cancer Institute, Pittsburgh, PA, United States
| | - Vincent Chung
- City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | | | - Ebenezer A Kio
- Goshen Center for Cancer Care, Goshen, IN, United States
| | | | - Roland T Skeel
- University of Toledo Medical Center, Toledo, OH, United States
| | - Olatunji B Alese
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Dana B Cardin
- Vanderbilt-Ingram Cancer Center, Nashville, TN, United States
| | | | - Wahid T Hanna
- University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Alexis D Leal
- University of Colorado School of Medicine, Aurora, CO, United States
| | - Valerie Lee
- Johns Hopkins University School of Medicine, Baltimore, MA, United States
| | - Anne M Noonan
- The Ohio State University, Columbus, OH, United States
| | - Philip A Philip
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Zev A Wainberg
- University of California School of Medicine, Los Angeles, CA, United States
| | - Hristina Pashova
- Corcept Therapeutics Incorporated, Menlo Park, CA, United States
| | - Grace Mann
- Corcept Therapeutics Incorporated, Menlo Park, CA, United States
| | | |
Collapse
|
2
|
Andres S, Finch L, Iasonos A, Zhou Q, Girshman J, Chhetri-Long R, Green H, Jang D, O'Cearbhaill R, Kyi C, Cohen S, Friedman C, Makker V, Chi DS, Sonoda Y, Chiang S, Aghajanian C, Weigelt B, Grisham RN. Basket study of oral progesterone antagonist onapristone extended release in progesterone receptor-positive recurrent granulosa cell, low-grade serous ovarian cancer, or endometrioid endometrial cancer. Gynecol Oncol 2024; 189:30-36. [PMID: 38991472 DOI: 10.1016/j.ygyno.2024.06.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/13/2024]
Abstract
OBJECTIVE To determine the safety and efficacy of the oral progesterone antagonist onapristone extended release (onapristone-XR) in patients with recurrent progesterone receptor (PR)-positive adult-type granulosa cell tumor (aGCT), low-grade serous ovarian cancer (LGSOC), or endometrioid endometrial cancer (EEC). METHODS This single-institution phase II study included patients with PR-positive aGCT, LGSOC, or EEC who received ≥1 prior line of chemotherapy. Patients were enrolled from 5/2019-5/2020. PR status was evaluated via immunohistochemistry. Eligible patients had PR expression ≥1% on tissue collected within 3 years of enrollment. Patients received 50 mg of onapristone-XR twice daily until disease progression or treatment discontinuation. Adverse events were graded by Common Terminology Criteria for Adverse Events version 5.0. The primary endpoint was overall response rate (ORR) by Response Evaluation Criteria in Solid Tumors 1.1. Secondary endpoints were response duration, clinical benefit rate (CBR), and safety. RESULTS Five patients with LGSOC and 1 with EEC enrolled, but both cohorts closed early due to slow accrual. Fourteen patients with aGCT enrolled and completed stage 1 accrual. No responses were observed. Four patients with LGSOC were evaluable, with median PFS of 4.4 months (range, 1.8-NE) and CBR of 50% (range, 6.8%-93.2%). All 14 patients with aGCT were evaluable, with median PFS of 2.8 months (range, 1.6-4.9), 6-month PFS rate of 21.4% (range, 5.2%-44.8%), 12-month PFS rate of 14.3% (range, 2.3%-36.6%), and a CBR of 35.7% (range, 12.8%-64.9%). CONCLUSIONS The study did not meet its primary endpoint. While onapristone-XR was well tolerated in all 3 arms, no objective responses were observed.
Collapse
MESH Headings
- Humans
- Female
- Middle Aged
- Aged
- Receptors, Progesterone/metabolism
- Endometrial Neoplasms/drug therapy
- Endometrial Neoplasms/pathology
- Neoplasm Recurrence, Local/drug therapy
- Carcinoma, Endometrioid/drug therapy
- Carcinoma, Endometrioid/pathology
- Carcinoma, Endometrioid/metabolism
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/metabolism
- Granulosa Cell Tumor/drug therapy
- Granulosa Cell Tumor/metabolism
- Granulosa Cell Tumor/pathology
- Adult
- Gonanes/administration & dosage
- Gonanes/adverse effects
- Delayed-Action Preparations/administration & dosage
- Aged, 80 and over
- Administration, Oral
- Cystadenocarcinoma, Serous/drug therapy
- Cystadenocarcinoma, Serous/pathology
- Cystadenocarcinoma, Serous/metabolism
Collapse
Affiliation(s)
- Sarah Andres
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lindsey Finch
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexia Iasonos
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Qin Zhou
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeffrey Girshman
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rashmi Chhetri-Long
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hunter Green
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dasom Jang
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Roisin O'Cearbhaill
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Chrisann Kyi
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Seth Cohen
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Claire Friedman
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Vicky Makker
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Dennis S Chi
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Yukio Sonoda
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Sarah Chiang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Carol Aghajanian
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rachel N Grisham
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
3
|
Kurkin DV, Bakupin DA, Morkovin EI, Krysanov IS, Makarova EV, Tsaplina AP, Klabukova DL, Ivanova OV, Gorbunova YV, Dzhavakhyan MA, Zvereva VI, Kolosov YA, Aleshnikova KY. Thalidomide: History of Research and Perspectives for Its Medical Use (Review). Pharm Chem J 2024; 58:1001-1010. [DOI: 10.1007/s11094-024-03236-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Indexed: 01/04/2025]
|
4
|
Giudice E, Salutari V, Sassu CM, Ghizzoni V, Carbone MV, Vertechy L, Fagotti A, Scambia G, Marchetti C. Relacorilant in recurrent ovarian cancer: clinical evidence and future perspectives. Expert Rev Anticancer Ther 2024; 24:649-655. [PMID: 38861580 DOI: 10.1080/14737140.2024.2362178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024]
Abstract
INTRODUCTION Relacorilant (CORT125134, Corcept Therapeutics) is a selective glucocorticoid receptor modulator, which reverses the glucocorticoid-mediated anti-apoptotic effects and restores the taxane chemosensitivity in epithelial ovarian cancer cells. Given those preclinical findings, relacorilant is currently under investigation in clinical trials in combination with nab-paclitaxel for the platinum-resistant ovarian cancer setting. AREAS COVERED Already published preclinical and clinical evidence of relacorilant antitumor activity was analyzed and discussed. Ongoing clinical trials registered on clincaltrials.gov were also reported. The review aimed to summarize the status of relacorilant, the mechanism of action, the published and ongoing trials, and its safety and efficacy. EXPERT OPINION Relacorilant combined with nab-paclitaxel, may represent a promising strategy for the treatment of platinum-resistant ovarian cancer patients. After preliminary positive results in terms of clinical efficacy, a randomized phase III trial is ongoing to confirm the findings from the published phase II study.
Collapse
Affiliation(s)
- Elena Giudice
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Vanda Salutari
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carolina Maria Sassu
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Viola Ghizzoni
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Maria Vittoria Carbone
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Laura Vertechy
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Anna Fagotti
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Institute of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Scambia
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Institute of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Claudia Marchetti
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Institute of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
5
|
Olawaiye AB, Kim JW, Bagameri A, Bishop E, Chudecka-Głaz A, Devaux A, Gladieff L, Gordinier ME, Korach J, McCollum ME, Mileshkin L, Monk BJ, Nicum S, Nogueira-Rodrigues A, Oaknin A, O'Malley DM, Orlando M, Dreiling L, Tudor IC, Lorusso D. Clinical Trial Protocol for ROSELLA: a phase 3 study of relacorilant in combination with nab-paclitaxel versus nab-paclitaxel monotherapy in advanced platinum-resistant ovarian cancer. J Gynecol Oncol 2024; 35:e111. [PMID: 39032926 PMCID: PMC11262895 DOI: 10.3802/jgo.2024.35.e111] [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: 05/28/2024] [Accepted: 06/11/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Ovarian cancer has the highest mortality among gynecologic cancers, primarily because it typically is diagnosed at a late stage and because of the development of chemoresistance in recurrent disease. Improving outcomes in women with platinum-resistant ovarian cancer is a substantial unmet need. Activation of the glucocorticoid receptor (GR) by cortisol has been shown to suppress the apoptotic pathways used by cytotoxic agents, limiting their efficacy. Selective GR modulation may be able to counteract cortisol's antiapoptotic effects, enhancing chemotherapy's efficacy. A previous phase 2 study has shown that adding intermittently dosed relacorilant, a selective GR modulator, to nab-paclitaxel improved outcomes, including progression-free survival (PFS) and overall survival (OS), with minimal added toxicity, in women with recurrent platinum-resistant ovarian cancer. The ROSELLA study aims to confirm and expand on these findings in a larger population. METHODS ROSELLA is a phase 3, randomized, 2-arm, open-label, global multicenter study in women with recurrent, platinum-resistant, high-grade serous epithelial ovarian, primary peritoneal, or fallopian tube cancer. Eligible participants have received 1 to 3 lines of prior systemic anticancer therapy, including ≥1 prior line of platinum therapy and prior treatment with bevacizumab, with documented progressive disease or intolerance to the most recent therapy. There is no biomarker-based requirement for participant selection. Participants are randomized 1:1 to receive intermittently dosed relacorilant in combination with nab-paclitaxel or nab-paclitaxel monotherapy. The study's primary efficacy endpoint is PFS as assessed by blinded independent central review. Secondary efficacy endpoints include OS, investigator-assessed PFS, objective response rate, best overall response, duration of response, clinical benefit rate at 24 weeks, and cancer antigen 125 response. The study is also evaluating safety and patient-reported outcomes. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05257408; European Union Drug Regulating Authorities Clinical Trials Database Identifier: 2022-000662-18.
Collapse
Affiliation(s)
- Alexander B Olawaiye
- University of Pittsburgh School of Medicine and Magee-Womens Hospital, Gynecologic Oncology Group, Pittsburgh, PA, USA.
| | - Jae-Weon Kim
- Department of Obstetrics and Gynecology, Seoul National University, Seoul, Korea
| | | | - Erin Bishop
- Medical College of Wisconsin, Gynecologic Oncology Group, Milwaukee, WI, USA
| | - Anita Chudecka-Głaz
- Pomeranian Medical University, Polish Gynecologic Oncology Group, Szczecin, Poland
| | - Alix Devaux
- Oncology Department of Grand Hôpital de Charleroi, Charleroi, Belgium
| | - Laurence Gladieff
- Institut Claudius Regaud-Institut Universitaire du Cancer de Toulouse Oncopole, Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens, Toulouse, France
| | | | - Jacob Korach
- Sheba Medical Center, School of Medicine, Tel Aviv University, Israeli Society of Gynecologic Oncology, Tel Aviv, Israel
| | | | - Linda Mileshkin
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Bradley J Monk
- Gynecologic Oncology Group Foundation; Florida Cancer Specialists and Research Institute, West Palm Beach, FL, USA
| | - Shibani Nicum
- University College London Cancer Institute, National Cancer Research Institute, London, UK
| | | | - Ana Oaknin
- Medical Oncology Service, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - David M O'Malley
- The Ohio State University and the James Cancer Center, Gynecologic Oncology Group, Columbus, OH, USA
| | - Mauro Orlando
- Instituto Alexander Fleming, Buenos Aires, Argentina
| | | | - Iulia C Tudor
- Corcept Therapeutics Incorporated, Menlo Park, CA, USA
| | - Domenica Lorusso
- Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico and Catholic University of the Sacred Heart, Multicentre Italian Trials in Ovarian Cancer and Gynecologic Malignancies, Rome, Italy
| |
Collapse
|
6
|
Taya M, Hou X, Veneris JT, Kazi N, Larson MC, Maurer MJ, Heinzen EP, Chen H, Lastra R, Oberg AL, Weroha SJ, Fleming GF, Conzen SD. Investigation of selective glucocorticoid receptor modulation in high-grade serous ovarian cancer PDX models. J Gynecol Oncol 2024; 36:36.e4. [PMID: 38909640 DOI: 10.3802/jgo.2025.36.e4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/18/2024] [Accepted: 05/07/2024] [Indexed: 06/25/2024] Open
Abstract
OBJECTIVE In ovarian cancer (OvCa), tumor cell high glucocorticoid receptor (GR) has been associated with poor patient prognosis. In vitro, GR activation inhibits chemotherapy-induced OvCa cell death in association with transcriptional upregulation of genes encoding anti-apoptotic proteins. A recent randomized phase II study demonstrated improvement in progression-free survival (PFS) for heavily pre-treated OvCa patients randomized to receive therapy with a selective GR modulator (SGRM) plus chemotherapy compared to chemotherapy alone. We hypothesized that SGRM therapy would improve carboplatin response in OvCa patient-derived xenograft (PDX). METHODS Six high-grade serous (HGS) OvCa PDX models expressing GR mRNA (NR3C1) and protein were treated with chemotherapy +/- SGRM. Tumor size was measured longitudinally by peritoneal transcutaneous ultrasonography. RESULTS One of the 6 GR-positive PDX models showed a significant improvement in PFS with the addition of a SGRM. Interestingly, the single model with an improved PFS was least carboplatin sensitive. Possible explanations for the modest SGRM activity include the high carboplatin sensitivity of 5 of the PDX tumors and the potential that SGRMs activate the tumor invasive immune cells in patients (absent from immunocompromised mice). The level of tumor GR protein expression alone appears insufficient for predicting SGRM response. CONCLUSION The significant improvement in PFS shown in 1 of the 6 models after treatment with a SGRM plus chemotherapy underscores the need to determine predictive biomarkers for SGRM therapy in HGS OvCa and to better identify patient subgroups that are most likely to benefit from adding GR modulation to chemotherapy.
Collapse
Affiliation(s)
- Manisha Taya
- Division of Hematology and Oncology, UT Southwestern, Dallas, TX, USA
| | - Xiaonan Hou
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Jennifer T Veneris
- Department of Medicine, Section of Hematology and Oncology, The University of Chicago, Chicago, IL, USA
| | - Nina Kazi
- Division of Hematology and Oncology, UT Southwestern, Dallas, TX, USA
| | - Melissa C Larson
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Matthew J Maurer
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Ethan P Heinzen
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, USA
| | - Hao Chen
- Department of Pathology, UT Southwestern, Dallas, TX, USA
| | - Ricardo Lastra
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Ann L Oberg
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - S John Weroha
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Gini F Fleming
- Department of Medicine, Section of Hematology and Oncology, The University of Chicago, Chicago, IL, USA
| | - Suzanne D Conzen
- Division of Hematology and Oncology, UT Southwestern, Dallas, TX, USA.
| |
Collapse
|
7
|
Yamazaki H, Bunbai K, Deguchi T, Tamura M, Ohota H. Comparison of chemotherapy outcomes between normal and high serum cortisol concentration in dogs with lymphoma. J Vet Intern Med 2024; 38:1651-1658. [PMID: 38471970 PMCID: PMC11099743 DOI: 10.1111/jvim.17044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Increased serum cortisol (COR) concentrations may induce glucocorticoid resistance by down-regulation of glucocorticoid receptor (GCR), resulting in decreased chemotherapy efficacy in dogs with lymphoma. HYPOTHESIS Investigate the relationship between serum COR concentrations and chemotherapy outcomes in dogs with lymphoma. ANIMALS Thirty client-owned dogs with lymphoma, with serum COR concentration measured using serum samples collected at diagnosis. METHODS Retrospective study. Dogs were divided into 2 groups based on serum COR concentrations: a normal group (n = 16) with COR concentrations <6 μg/dL and a high group (14) with COR concentrations ≥6 μg/dL. We compared signalment, clinical signs, stage, type of lymphoma, adrenal gland size, alkaline phosphatase (ALP) activity, response to chemotherapy, progression-free survival (PFS), overall survival (OS), and rate of P-glycoprotein (P-gp)- and GCR-positive cells between the 2 groups. RESULTS No significant differences were found in the demographic characteristics between the 2 groups. However, the high COR group exhibited a significantly lower response to chemotherapy, PFS, and OS compared with the normal COR group. Serum ALP activity was significantly higher in the high COR group than in the normal COR group. Adrenal gland size was also significantly larger in the high COR group. Although no significant differences were found in the rate of P-gp-positive cells between the 2 groups, the rate of GCR-positive cells was significantly lower in the high COR group. CONCLUSIONS AND CLINICAL IMPORTANCE Our data suggests that measurement of serum COR concentrations may serve as a potential prognostic factor and evaluation index.
Collapse
Affiliation(s)
- Hiroki Yamazaki
- Laboratory of Companion Animal Internal Medicine, Department of Companion Animal, Clinical Sciences, School of Veterinary MedicineRakuno Gakuen UniversityEbetsuJapan
| | - Kaito Bunbai
- Laboratory of Companion Animal Internal Medicine, Department of Companion Animal, Clinical Sciences, School of Veterinary MedicineRakuno Gakuen UniversityEbetsuJapan
| | - Tatsuya Deguchi
- Laboratory of Companion Animal Internal Medicine, Department of Companion Animal, Clinical Sciences, School of Veterinary MedicineRakuno Gakuen UniversityEbetsuJapan
| | - Masahiro Tamura
- Laboratory of Companion Animal Internal Medicine, Department of Companion Animal, Clinical Sciences, School of Veterinary MedicineRakuno Gakuen UniversityEbetsuJapan
| | - Hiroshi Ohota
- Laboratory of Companion Animal Internal Medicine, Department of Companion Animal, Clinical Sciences, School of Veterinary MedicineRakuno Gakuen UniversityEbetsuJapan
| |
Collapse
|
8
|
Rocha SM, Gustafson DL, Safe S, Tjalkens RB. Comparative safety, pharmacokinetics, and off-target assessment of 1,1-bis(3'-indolyl)-1-( p-chlorophenyl) methane in mouse and dog: implications for therapeutic development. Toxicol Res (Camb) 2024; 13:tfae059. [PMID: 38655145 PMCID: PMC11033559 DOI: 10.1093/toxres/tfae059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/26/2024] Open
Abstract
The modified phytochemical derivative, 1,1-bis(3'-indolyl)-1-(p-chlorophenyl) methane (C-DIM12), has been identified as a potential therapeutic platform based on its capacity to improve disease outcomes in models of neurodegeneration and cancer. However, comprehensive safety studies investigating pathology and off-target binding have not been conducted. To address this, we administered C-DIM12 orogastrically to outbred male CD-1 mice for 7 days (50 mg/kg/day, 200 mg/kg/day, and 300 mg/kg/day) and investigated changes in hematology, clinical chemistry, and whole-body tissue pathology. We also delivered a single dose of C-DIM12 (1 mg/kg, 5 mg/kg, 25 mg/kg, 100 mg/kg, 300 mg/kg, 1,000 mg/kg) orogastrically to male and female beagle dogs and investigated hematology and clinical chemistry, as well as plasma pharmacokinetics over 48-h. Consecutive in-vitro off-target binding through inhibition was performed with 10 μM C-DIM12 against 68 targets in tandem with predictive off-target structural binding capacity. These data show that the highest dose C-DIM12 administered in each species caused modest liver pathology in mouse and dog, whereas lower doses were unremarkable. Off-target screening and predictive modeling of C-DIM12 show inhibition of serine/threonine kinases, calcium signaling, G-protein coupled receptors, extracellular matrix degradation, and vascular and transcriptional regulation pathways. Collectively, these data demonstrate that low doses of C-DIM12 do not induce pathology and are capable of modulating targets relevant to neurodegeneration and cancer.
Collapse
Affiliation(s)
- Savannah M Rocha
- Department of Environmental and Radiological Health Sciences, Colorado State University, 1680 Campus Delivery Fort Collins, CO 80523, USA
| | - Daniel L Gustafson
- Department of Clinical Sciences, Colorado State University, 1678 Campus Delivery Fort Collins, CO 80523, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M School of Veterinary, Medicine & Biomedical Sciences, 4466 TAMU College Station, TX 77843-4466, USA
| | - Ronald B Tjalkens
- Department of Environmental and Radiological Health Sciences, Colorado State University, 1680 Campus Delivery Fort Collins, CO 80523, USA
| |
Collapse
|
9
|
Wen X, Xiao Y, Xiao H, Tan X, Wu B, Li Z, Wang R, Xu X, Li T. Bisphenol S induces brown adipose tissue whitening and aggravates diet-induced obesity in an estrogen-dependent manner. Cell Rep 2023; 42:113504. [PMID: 38041811 DOI: 10.1016/j.celrep.2023.113504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/06/2023] [Accepted: 11/10/2023] [Indexed: 12/04/2023] Open
Abstract
Bisphenol S (BPS) exposure has been implied epidemiologically to increase obesity risk, but the underlying mechanism is unclear. Here, we propose that BPS exposure at an environmentally relevant dose aggravates diet-induced obesity in female mice by inducing brown adipose tissue (BAT) whitening. We explored the underlying mechanism by which KDM5A-associated demethylation of the trimethylation of lysine 4 on histone H3 (H3K4me3) in thermogenic genes is overactivated in BAT upon BPS exposure, leading to the reduced expression of thermogenic genes. Further studies have suggested that BPS activates KDM5A transcription in BAT by binding to glucocorticoid receptor (GR) in an estrogen-dependent manner. Estrogen-estrogen receptors facilitate the accessibility of the KDM5A gene promoter to BPS-activated GR by recruiting the activator protein 1 (AP-1) complex. These results indicate that BAT is another important target of BPS and that targeting KDM5A-related signals may serve as an approach to counteract the BPS-induced susceptivity to obesity.
Collapse
Affiliation(s)
- Xue Wen
- Department of Plastic and Burn Surgery, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China; Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yang Xiao
- Department of Plastic and Burn Surgery, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Haitao Xiao
- Department of Plastic and Burn Surgery, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xueqin Tan
- Department of Plastic and Burn Surgery, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China; Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Beiyi Wu
- Department of Plastic and Burn Surgery, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China; Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Zehua Li
- Department of Plastic and Burn Surgery, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China; Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ru Wang
- Department of Plastic and Burn Surgery, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xuewen Xu
- Department of Plastic and Burn Surgery, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China.
| | - Tao Li
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu 610041, China.
| |
Collapse
|
10
|
Colombo N, Van Gorp T, Matulonis UA, Oaknin A, Grisham RN, Fleming GF, Olawaiye AB, Nguyen DD, Greenstein AE, Custodio JM, Pashova HI, Tudor IC, Lorusso D. Relacorilant + Nab-Paclitaxel in Patients With Recurrent, Platinum-Resistant Ovarian Cancer: A Three-Arm, Randomized, Controlled, Open-Label Phase II Study. J Clin Oncol 2023; 41:4779-4789. [PMID: 37364223 PMCID: PMC10602497 DOI: 10.1200/jco.22.02624] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 04/21/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
PURPOSE Despite therapeutic advances, outcomes for patients with platinum-resistant/refractory ovarian cancer remain poor. Selective glucocorticoid receptor modulation with relacorilant may restore chemosensitivity and enhance chemotherapy efficacy. METHODS This three-arm, randomized, controlled, open-label phase II study (ClinicalTrials.gov identifier: NCT03776812) enrolled women with recurrent, platinum-resistant/refractory, high-grade serous or endometrioid epithelial ovarian, primary peritoneal, or fallopian tube cancer, or ovarian carcinosarcoma treated with ≤4 prior chemotherapeutic regimens. Patients were randomly assigned 1:1:1 to (1) nab-paclitaxel (80 mg/m2) + intermittent relacorilant (150 mg the day before, of, and after nab-paclitaxel); (2) nab-paclitaxel (80 mg/m2) + continuous relacorilant (100 mg once daily); or (3) nab-paclitaxel monotherapy (100 mg/m2). Nab-paclitaxel was administered on days 1, 8, and 15 of each 28-day cycle. The primary end point was progression-free survival (PFS) by investigator assessment; objective response rate (ORR), duration of response (DOR), overall survival (OS), and safety were secondary end points. RESULTS A total of 178 women were randomly assigned. Intermittent relacorilant + nab-paclitaxel improved PFS (hazard ratio [HR], 0.66; log-rank test P = .038; median follow-up, 11.1 months) and DOR (HR, 0.36; P = .006) versus nab-paclitaxel monotherapy, while ORR was similar across arms. At the preplanned OS analysis (median follow-up, 22.5 months), the OS HR was 0.67 (P = .066) for the intermittent arm versus nab-paclitaxel monotherapy. Continuous relacorilant + nab-paclitaxel showed numerically improved median PFS but did not result in significant improvement over nab-paclitaxel monotherapy. Adverse events were comparable across study arms, with neutropenia, anemia, peripheral neuropathy, and fatigue/asthenia being the most common grade ≥3 adverse events. CONCLUSION Intermittent relacorilant + nab-paclitaxel improved PFS, DOR, and OS compared with nab-paclitaxel monotherapy. On the basis of protocol-prespecified Hochberg step-up multiplicity adjustment, the primary end point did not reach statistical significance (P < .025). A phase III evaluation of this regimen is underway (ClinicalTrials.gov identifier: NCT05257408).
Collapse
Affiliation(s)
- Nicoletta Colombo
- Gynecologic Oncology Program, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Medicine and Surgery, University Milan-Bicocca, Milan, Italy
| | - Toon Van Gorp
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University Hospital Leuven, Leuven Cancer Institute, Leuven, Belgium
| | | | - Ana Oaknin
- Gynaecologic Cancer Programme, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Rachel N. Grisham
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical Center, New York, NY
| | | | - Alexander B. Olawaiye
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | | | | | | | | | - Domenica Lorusso
- Fondazione Policlinico Universitario Gemelli IRCCS, Catholic University of Sacred Heart, Rome, Italy
| |
Collapse
|
11
|
Johnson KCC, Goldstein D, Tharakan J, Quiroga D, Kassem M, Grimm M, Miah A, Vargo C, Berger M, Sudheendra P, Pariser A, Gatti-Mays ME, Williams N, Stover D, Sardesai S, Wesolowski R, Ramaswamy B, Tozbikian G, Schnell PM, Cherian MA. The Immunomodulatory Effects of Dexamethasone on Neoadjuvant Chemotherapy for Triple-Negative Breast Cancer. Oncol Ther 2023; 11:361-374. [PMID: 37354381 PMCID: PMC10447758 DOI: 10.1007/s40487-023-00235-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 05/26/2023] [Indexed: 06/26/2023] Open
Abstract
INTRODUCTION The immunomodulatory impact of corticosteroids and concurrent chemotherapy is poorly understood within triple-negative breast cancer (TNBC). On a biochemical level, steroids have been linked to the signaling of chemotherapy-resistant pathways. However, on a clinical level, steroids play an essential role in chemotherapy tolerance through the prevention of chemotherapy-induced nausea and vomiting (CINV) and hypersensitivity reactions. Given these conflicting roles, we wanted to evaluate this interplay more rigorously in the context of early-stage TNBC. METHODS We performed a retrospective analysis of patients with operable TNBC who received neoadjuvant chemotherapy (NAC) between January 2012 and November 2018, with the primary goal of examining the dose-dependent relationship between pathological complete response (pCR) rates and corticosteroid use. Secondary endpoints included the impact of steroid dosing on overall survival (OS) and recurrence-free survival (RFS), along with a breakdown in pCR rates based on steroid doses provided during each chemotherapy phase. Further adjusted analyses were performed based on patient age, diabetic status, and anatomical stage. Finally, we explored the relationship between tumor-infiltrating lymphocytes (TILs) seen on tissue samples at baseline and dexamethasone doses in terms of pCR rates. RESULTS In total, of the 174 patients screened within this study period, 116 met full eligibility criteria. Of these eligible patients, all were female, with a median age of 51.5 years (27.0 to 74.0) and a mean body mass index (BMI) of 29.7 [standard deviation (SD) 7.04]. The majority were nondiabetic (80.2%). For cancer stage, 69.8% (n = 81) had stage 2 breast cancer. We found no statistically significant association between pCR rates and dexamethasone use, both in terms of the total dose (p = 0.55) and mean dose per NAC cycle (p = 0.74). Similarly, no difference was noted when adjusting for diabetic status, metformin use, or age at diagnosis, regardless of the total steroid dose provided (p = 0.72) or mean dose per cycle (p = 0.49). No meaningful changes to pCR rate were seen with higher mean or higher total steroid doses during the paclitaxel (T) phase (adjusted p = 0.16 and p = 0.76, respectively) or doxorubicin and cyclophosphamide (AC) phase (adjusted p = 0.83 and p = 0.77, respectively). Furthermore, we found no clinically significant association between dexamethasone dose and either RFS (p = 0.45) or OS (p = 0.89). Of the 56 patients who had available pre-treatment biopsy tissue samples, 27 achieved pCR, with higher TILs at baseline being associated with higher pCR rates, regardless of the mean dexamethasone dose used. CONCLUSION Our findings demonstrate that dexamethasone has no clinically significant impact on pCR, RFS, or OS when given concurrently with NAC in patients with curative TNBC, regardless of diabetic status.
Collapse
Affiliation(s)
- Kai Conrad Cecil Johnson
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | | | - Jasmin Tharakan
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Dionisia Quiroga
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Mahmoud Kassem
- Department of Surgery, Mercy Health West Hospital, Cincinnati, OH, USA
| | - Michael Grimm
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Abdul Miah
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Craig Vargo
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Michael Berger
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Preeti Sudheendra
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Ashley Pariser
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Margaret E Gatti-Mays
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Nicole Williams
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Daniel Stover
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Sagar Sardesai
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Robert Wesolowski
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Bhuvaneswari Ramaswamy
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Gary Tozbikian
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Patrick M Schnell
- Division of Biostatistics, The Ohio State University College of Public Health, Columbus, OH, USA
| | - Mathew A Cherian
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
12
|
Sanchez-Hernandez ES, Ochoa PT, Suzuki T, Ortiz-Hernandez GL, Unternaehrer JJ, Alkashgari HR, Diaz Osterman CJ, Martinez SR, Chen Z, Kremsky I, Wang C, Casiano CA. Glucocorticoid Receptor Regulates and Interacts with LEDGF/p75 to Promote Docetaxel Resistance in Prostate Cancer Cells. Cells 2023; 12:2046. [PMID: 37626856 PMCID: PMC10453226 DOI: 10.3390/cells12162046] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/31/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Patients with advanced prostate cancer (PCa) invariably develop resistance to anti-androgen therapy and taxane-based chemotherapy. Glucocorticoid receptor (GR) has been implicated in PCa therapy resistance; however, the mechanisms underlying GR-mediated chemoresistance remain unclear. Lens epithelium-derived growth factor p75 (LEDGF/p75, also known as PSIP1 and DFS70) is a glucocorticoid-induced transcription co-activator implicated in cancer chemoresistance. We investigated the contribution of the GR-LEDGF/p75 axis to docetaxel (DTX)-resistance in PCa cells. GR silencing in DTX-sensitive and -resistant PCa cells decreased LEDGF/p75 expression, and GR upregulation in enzalutamide-resistant cells correlated with increased LEDGF/p75 expression. ChIP-sequencing revealed GR binding sites in the LEDGF/p75 promoter. STRING protein-protein interaction analysis indicated that GR and LEDGF/p75 belong to the same transcriptional network, and immunochemical studies demonstrated their co-immunoprecipitation and co-localization in DTX-resistant cells. The GR modulators exicorilant and relacorilant increased the sensitivity of chemoresistant PCa cells to DTX-induced cell death, and this effect was more pronounced upon LEDGF/p75 silencing. RNA-sequencing of DTX-resistant cells with GR or LEDGF/p75 knockdown revealed a transcriptomic overlap targeting signaling pathways associated with cell survival and proliferation, cancer, and therapy resistance. These studies implicate the GR-LEDGF/p75 axis in PCa therapy resistance and provide a pre-clinical rationale for developing novel therapeutic strategies for advanced PCa.
Collapse
Affiliation(s)
- Evelyn S. Sanchez-Hernandez
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
| | - Pedro T. Ochoa
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
| | - Tise Suzuki
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
| | - Greisha L. Ortiz-Hernandez
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
| | - Juli J. Unternaehrer
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
| | - Hossam R. Alkashgari
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
- Department of Physiology, College of Medicine, University of Jeddah, Jeddah 23890, Saudi Arabia
| | - Carlos J. Diaz Osterman
- Department of Basic Sciences, Ponce Health Sciences University, Ponce, PR 00716, USA; (C.J.D.O.); (S.R.M.)
| | - Shannalee R. Martinez
- Department of Basic Sciences, Ponce Health Sciences University, Ponce, PR 00716, USA; (C.J.D.O.); (S.R.M.)
| | - Zhong Chen
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
- Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Isaac Kremsky
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
- Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Charles Wang
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
- Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Carlos A. Casiano
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
- Rheumatology Division, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
13
|
Mallick S, Chakrabarti J, Eschbacher J, Moraitis AG, Greenstein AE, Churko J, Pond KW, Livolsi A, Thorne CA, Little AS, Yuen KCJ, Zavros Y. Genetically engineered human pituitary corticotroph tumor organoids exhibit divergent responses to glucocorticoid receptor modulators. Transl Res 2023; 256:56-72. [PMID: 36640905 PMCID: PMC11345864 DOI: 10.1016/j.trsl.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/12/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023]
Abstract
Cushing's disease (CD) is a serious endocrine disorder attributed to an adrenocorticotropic hormone (ACTH)-secreting pituitary neuroendocrine tumor (PitNET) that that subsequently leads to chronic hypercortisolemia. PitNET regression has been reported following treatment with the investigational selective glucocorticoid receptor (GR) modulator relacorilant, but the mechanisms behind that effect remain unknown. Human PitNET organoid models were generated from induced human pluripotent stem cells (iPSCs) or fresh tissue obtained from CD patient PitNETs (hPITOs). Genetically engineered iPSC derived organoids were used to model the development of corticotroph PitNETs expressing USP48 (iPSCUSP48) or USP8 (iPSCUSP8) somatic mutations. Organoids were treated with the GR antagonist mifepristone or the GR modulator relacorilant with or without somatostatin receptor (SSTR) agonists pasireotide or octreotide. In iPSCUSP48 and iPSCUSP8 cultures, mifepristone induced a predominant expression of SSTR2 with a concomitant increase in ACTH secretion and tumor cell proliferation. Relacorilant predominantly induced SSTR5 expression and tumor cell apoptosis with minimal ACTH induction. Hedgehog signaling mediated the induction of SSTR2 and SSTR5 in response to mifepristone and relacorilant. Relacorilant sensitized PitNET organoid responsiveness to pasireotide. Therefore, our study identified the potential therapeutic use of relacorilant in combination with somatostatin analogs and demonstrated the advantages of relacorilant over mifepristone, supporting its further development for use in the treatment of Cushing's disease patients.
Collapse
Affiliation(s)
- Saptarshi Mallick
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Jayati Chakrabarti
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Jennifer Eschbacher
- Department of Neuropathology, Barrow Neurological Institute, Phoenix, Arizona
| | | | | | - Jared Churko
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Kelvin W Pond
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | | | - Curtis A Thorne
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Andrew S Little
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, Arizona
| | - Kevin C J Yuen
- Department of Neuroendocrinology, Barrow Neurological Institute, Phoenix, Arizona
| | - Yana Zavros
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona.
| |
Collapse
|
14
|
Greenstein AE, Hunt HJ. The glucocorticoid receptor modulator relacorilant reverses the immunosuppressive effects of cortisol. Int Immunopharmacol 2023; 120:110312. [PMID: 37230031 DOI: 10.1016/j.intimp.2023.110312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/26/2023] [Accepted: 05/07/2023] [Indexed: 05/27/2023]
Abstract
Cortisol, an endogenous glucocorticoid receptor (GR) agonist, controls a broad transcriptional program that affects T-cell activation, pro-inflammatory cytokine secretion, apoptosis, and immune-cell trafficking. The degree to which endogenous cortisol blunts the anti-tumor immune response checkpoint inhibitors stimulate had not been assessed. We addressed this question using relacorilant, a selective GR modulator (SGRM) that competitively antagonizes the effects of cortisol activity. GR expression in human tumor and immune cells positively correlated with PD-L1 expression and tumor infiltration of Th2 and Treg cells, and negatively correlated with Th1-cell infiltration. In vitro, cortisol inhibited, and relacorilant restored, T-cell activation and pro-inflammatory cytokine secretion in human peripheral blood mononuclear cells. In the ovalbumin-expressing EG7 and MC38 immune-competent tumor models, relacorilant significantly improved anti-PD-1 antibody efficacy and showed favorable effects on antigen-specific T-cells and systemic TNFα and IL-10. These data characterize the broad immunosuppressive effects of endogenous cortisol and highlight the potential of combining an SGRM with an immune checkpoint inhibitor.
Collapse
Affiliation(s)
| | - Hazel J Hunt
- Corcept Therapeutics, 149 Commonwealth Dr, Menlo Park, CA 94025, USA.
| |
Collapse
|
15
|
Buonaiuto R, Neola G, Cecere SC, Caltavituro A, Cefaliello A, Pietroluongo E, De Placido P, Giuliano M, Arpino G, De Angelis C. Glucocorticoid Receptor and Ovarian Cancer: From Biology to Therapeutic Intervention. Biomolecules 2023; 13:biom13040653. [PMID: 37189400 DOI: 10.3390/biom13040653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023] Open
Abstract
Ovarian cancer (OC) is the leading cause of death from gynecological malignancies worldwide. Fortunately, recent advances in OC biology and the discovery of novel therapeutic targets have led to the development of novel therapeutic agents that may improve the outcome of OC patients. The glucocorticoid receptor (GR) is a ligand-dependent transcriptional factor known for its role in body stress reactions, energy homeostasis and immune regulation. Notably, evidence suggests that GR may play a relevant role in tumor progression and may affect treatment response. In cell culture models, administration of low levels of glucocorticoids (GCs) suppresses OC growth and metastasis. Conversely, high GR expression has been associated with poor prognostic features and long-term outcomes in patients with OC. Moreover, both preclinical and clinical data have shown that GR activation impairs the effectiveness of chemotherapy by inducing the apoptotic pathways and cell differentiation. In this narrative review, we summarize data related to the function and role of GR in OC. To this aim, we reorganized the controversial and fragmented data regarding GR activity in OC and herein describe its potential use as a prognostic and predictive biomarker. Moreover, we explored the interplay between GR and BRCA expression and reviewed the latest therapeutic strategies such as non-selective GR antagonists and selective GR modulators to enhance chemotherapy sensitivity, and to finally provide new treatment options in OC patients.
Collapse
Affiliation(s)
- Roberto Buonaiuto
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Giuseppe Neola
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Sabrina Chiara Cecere
- Oncologia Clinica Sperimentale Uro-Ginecologica, Istituto Nazionale Tumori IRCCS Fondazione G Pascale, 80131 Naples, Italy
| | - Aldo Caltavituro
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Amedeo Cefaliello
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Erica Pietroluongo
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Pietro De Placido
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
16
|
Obrador E, Salvador-Palmer R, López-Blanch R, Oriol-Caballo M, Moreno-Murciano P, Estrela JM. Survival Mechanisms of Metastatic Melanoma Cells: The Link between Glucocorticoids and the Nrf2-Dependent Antioxidant Defense System. Cells 2023; 12:cells12030418. [PMID: 36766760 PMCID: PMC9913432 DOI: 10.3390/cells12030418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/11/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
Circulating glucocorticoids increase during stress. Chronic stress, characterized by a sustained increase in serum levels of cortisol, has been associated in different cases with an increased risk of cancer and a worse prognosis. Glucocorticoids can promote gluconeogenesis, mobilization of amino acids, fat breakdown, and impair the body's immune response. Therefore, conditions that may favor cancer growth and the acquisition of radio- and chemo-resistance. We found that glucocorticoid receptor knockdown diminishes the antioxidant protection of murine B16-F10 (highly metastatic) melanoma cells, thus leading to a drastic decrease in their survival during interaction with the vascular endothelium. The BRAFV600E mutation is the most commonly observed in melanoma patients. Recent studies revealed that VMF/PLX40-32 (vemurafenib, a selective inhibitor of mutant BRAFV600E) increases mitochondrial respiration and reactive oxygen species (ROS) production in BRAFV600E human melanoma cell lines. Early-stage cancer cells lacking Nrf2 generate high ROS levels and exhibit a senescence-like growth arrest. Thus, it is likely that a glucocorticoid receptor antagonist (RU486) could increase the efficacy of BRAF-related therapy in BRAFV600E-mutated melanoma. In fact, during early progression of skin melanoma metastases, RU486 and VMF induced metastases regression. However, treatment at an advanced stage of growth found resistance to RU486 and VMF. This resistance was mechanistically linked to overexpression of proteins of the Bcl-2 family (Bcl-xL and Mcl-1 in different human models). Moreover, melanoma resistance was decreased if AKT and NF-κB signaling pathways were blocked. These findings highlight mechanisms by which metastatic melanoma cells adapt to survive and could help in the development of most effective therapeutic strategies.
Collapse
Affiliation(s)
- Elena Obrador
- Cell Pathophysiology Unit (UFC), Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
- Scientia BioTech S.L., 46002 Valencia, Spain
- Correspondence: (E.O.); (J.M.E.); Tel.: +34-963864646 (J.M.E.)
| | - Rosario Salvador-Palmer
- Cell Pathophysiology Unit (UFC), Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
| | - Rafael López-Blanch
- Cell Pathophysiology Unit (UFC), Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
- Scientia BioTech S.L., 46002 Valencia, Spain
| | - María Oriol-Caballo
- Cell Pathophysiology Unit (UFC), Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
- Scientia BioTech S.L., 46002 Valencia, Spain
| | | | - José M. Estrela
- Cell Pathophysiology Unit (UFC), Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
- Scientia BioTech S.L., 46002 Valencia, Spain
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
- Correspondence: (E.O.); (J.M.E.); Tel.: +34-963864646 (J.M.E.)
| |
Collapse
|
17
|
Blanco-Nistal MM, Fernández-Fernández JA. Glucocorticoid Effect in Cancer Patients. Methods Mol Biol 2023; 2704:339-352. [PMID: 37642855 DOI: 10.1007/978-1-0716-3385-4_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
The use of glucocorticoids is very varied in the context of cancer patients and includes the treatment of symptoms related to cancer, but also the management of the most common side effects of antitumor treatments or adverse events related to the immune system. There is a quantity of experimental evidence demonstrating that cancer cells are immunogenic. However, the effective activation of anticancer T cell responses closely depends on an efficient antigen presentation carried out by professional antigen-presenting cells such as dendritic cells (DCs). The classic strategies to improve the medical management of inflammation are aimed at exacerbating the host's immune response. Although successful in treating a number of diseases, these drugs have limited efficacy and variable responses can lead to unpredictable results. The ideal therapy should reduce inflammation without inducing immunosuppression and remains a challenge for healthcare personnel.
Collapse
|
18
|
Munster PN, Greenstein AE, Fleming GF, Borazanci E, Sharma MR, Custodio JM, Tudor IC, Pashova HI, Shepherd SP, Grauer A, Sachdev JC. Overcoming Taxane Resistance: Preclinical and Phase 1 Studies of Relacorilant, a Selective Glucocorticoid Receptor Modulator, with Nab-Paclitaxel in Solid Tumors. Clin Cancer Res 2022; 28:3214-3224. [PMID: 35583817 PMCID: PMC9662918 DOI: 10.1158/1078-0432.ccr-21-4363] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 04/01/2022] [Accepted: 05/16/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE Chemotherapy resistance remains a major problem in many solid tumors, including breast, ovarian, and pancreatic cancer. Glucocorticoids are one potential driver of chemotherapy resistance as they can mediate tumor progression via induction of cell-survival pathways. We investigated whether combining the selective glucocorticoid receptor (GR) modulator relacorilant with taxanes can enhance antitumor activity. PATIENTS AND METHODS The effect of relacorilant on paclitaxel efficacy was assessed in OVCAR5 cells in vitro and in the MIA PaCa-2 xenograft. A phase 1 study of patients with advanced solid tumors was conducted to determine the recommended phase 2 dose of relacorilant + nab-paclitaxel. RESULTS In OVCAR5 cells, relacorilant reversed the deleterious effects of glucocorticoids on paclitaxel efficacy (P < 0.001). Compared with paclitaxel alone, relacorilant + paclitaxel reduced tumor growth and slowed time to progression in xenograft models (both P < 0.0001). In the heavily pretreated phase 1 population [median (range) of prior regimens: 3 (1-8), prior taxane in 75.3% (55/73)], 33% (19/57) of response-evaluable patients achieved durable disease control (≥16 weeks) with relacorilant + nab-paclitaxel and 28.6% (12/42) experienced longer duration of benefit than on prior taxane (up to 6.4×). The most common dose-limiting toxicity of the combination was neutropenia, which was manageable with prophylactic G-CSF. Clinical benefit with relacorilant + nab-paclitaxel was also associated with GR-regulated transcript-level changes in a panel of GR-controlled genes. CONCLUSIONS The observed preclinical, clinical, and GR-specific pharmacodynamic responses demonstrate that selective GR modulation with relacorilant combined with nab-paclitaxel may promote chemotherapy response and is tolerable. Further evaluation of this combination in tumor types responsive to taxanes is ongoing.
Collapse
Affiliation(s)
- Pamela N. Munster
- Department of Medicine (Hematology/Oncology), University of California San Francisco, San Francisco, California.,Corresponding Author: Pamela N. Munster, University of California, San Francisco, Box 1711, San Francisco, CA 94143. Phone: 415-502-3414; E-mail:
| | | | - Gini F. Fleming
- Department of Medicine, the University of Chicago, Chicago, Illinois
| | | | - Manish R. Sharma
- Department of Medicine, the University of Chicago, Chicago, Illinois
| | | | | | | | | | | | | |
Collapse
|
19
|
High glucocorticoid receptor expression in the sarcomatous versus carcinomatous elements of Mullerian carcinosarcomas. Gynecol Oncol Rep 2022; 41:100987. [PMID: 35519002 PMCID: PMC9065886 DOI: 10.1016/j.gore.2022.100987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/21/2022] [Accepted: 04/24/2022] [Indexed: 11/23/2022] Open
|
20
|
Dorman K, Heinemann V, Kobold S, von Bergwelt-Baildon M, Boeck S. Novel systemic treatment approaches for metastatic pancreatic cancer. Expert Opin Investig Drugs 2022; 31:249-262. [PMID: 35114868 DOI: 10.1080/13543784.2022.2037552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) has a 5-year overall survival rate of 10 %, emphasizing the need for more effective therapies, especially in metastatic disease. The immunosuppressive tumor microenvironment, poor vascularization, and dense tumor stroma typical for PDAC are hurdles that need to be overcome by novel drugs. Investigations are moving towards more targeted treatments including immunotherapy and cell-based approaches. AREAS COVERED This article reviews emerging drugs in clinical development for metastatic PDAC, focusing on cellular therapies and novel treatments targeting metabolism, tumor stroma, oncogenic pathways and immunosuppression. With immunotherapy and CAR T cell therapy on the rise in hematological malignancies, the transfer to solid tumors remains intriguing. Multiple exciting clinical trials investigating innovative therapeutic strategies for PDAC are currently ongoing and reviewed herein. ClinicalTrials.gov, conference abstracts and PubMed were searched in August 2021 and assessed for information on ongoing and published clinical studies. EXPERT OPINION With many challenges to overcome, the optimal therapy for patients with metastatic PDAC is likely to consist of a combination of different agents. We are slowly moving from entity-dependent approaches to ones more focused on molecular and pathological features. Increasingly personalized treatment plans tailored to each patient may be the future of PDAC therapy.
Collapse
Affiliation(s)
- Klara Dorman
- Department of Internal Medicine III and Comprehensive Cancer Center, Klinikum Grosshadern, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Volker Heinemann
- Department of Internal Medicine III and Comprehensive Cancer Center, Klinikum Grosshadern, Ludwig-Maximilians-Universität München, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Sebastian Kobold
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,Center for Integrated Protein Science Munich and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Internal Medicine III and Comprehensive Cancer Center, Klinikum Grosshadern, Ludwig-Maximilians-Universität München, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Stefan Boeck
- Department of Internal Medicine III and Comprehensive Cancer Center, Klinikum Grosshadern, Ludwig-Maximilians-Universität München, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| |
Collapse
|