129 results on '"Ralph J. Hauke"'
Search Results
2. 665 Immunotherapeutic trial in men with biochemical recurrence after definitive local therapy for prostate cancer: A clinical trial in progress
- Author
-
Neal Shore, Robert Dreicer, Matthew Zibelman, Mark N Stein, Ralph J Hauke, Russell Pachynski, Bethan Jones, Jessica Hawley, Charlotte Davis, Katie Anderson, Antonella Vardeu, Margaret A Marshall, Vicky Wheeler, Sarah Sebastian, Kevin Kayvan Zarrabi, Brian Miles, and Jennifer Bendall
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
3. 742 Phase 1/2 study of the bispecific 4–1BB and PD-L1 antibody INBRX-105 alone and in combination with pembrolizumab in select solid tumors
- Author
-
David Hong, Manish Sharma, Erminia Massarelli, Heather Kinkead, Naomi B Haas, Rachel E Sanborn, Ralph J Hauke, Anthony W Tolcher, Neal Akhave, Jong Chul Park, Jennifer Carlisle, John Hamm, Alexander I Spira, David Berz, Vasily Andrianov, Brianne O’Neill, Justin A Call, Frank Yung-Chin Tsai, and D Ross Camidge
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
4. 748 Phase 1/2 study of the hexavalent OX40 agonist INBRX-106 alone and in combination with pembrolizumab in select solid tumors
- Author
-
Daniel Olson, Manish Sharma, Erminia Massarelli, Jonathan Thompson, Rachel E Sanborn, Ralph J Hauke, Anthony W Tolcher, Elizabeth Davis, Conor Steuer, Muhammad Furqan, John Hamm, Doug Laux, David Berz, Wade T Iams, Vasily Andrianov, Brianne O’Neill, and Yaiza Diaz De Durana
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
5. SALVO: Single-Arm Trial of Ipilimumab and Nivolumab as Adjuvant Therapy for Resected Mucosal Melanoma
- Author
-
Lisa A. Kottschade, Gregory Russell Pond, Anthony J. Olszanski, Yousef Zakharia, Evidio Domingo-Musibay, Ralph J. Hauke, Brendan D. Curti, Sarah Schober, Mohammed M. Milhem, Matthew Stephen Block, Tina Hieken, and Robert R. McWilliams
- Subjects
Cancer Research ,Oncology - Abstract
Purpose: Mucosal melanoma is a rare, aggressive form of melanoma with extremely high recurrence rates despite definitive surgical resection with curative intent. Currently there is no consensus on adjuvant therapy. Data on checkpoint inhibitors for adjuvant therapy are lacking. Patients and Methods: We performed a single-arm, multicenter clinical trial using “flip dose” ipilimumab (1 mg/kg q3w × 4 cycles), and nivolumab (3 mg/kg q3w × 4 cycles), then nivolumab 480 mg q4w × 11 cycles to complete a year of adjuvant therapy. Participants must have had R0/R1 resection ≤90 days before registration, no prior systemic therapy (adjuvant radiotherapy allowed), ECOG 0/1, and no uncontrolled autoimmune disease or other invasive cancer. Patients were recruited through the Midwest Melanoma Partnership/Hoosier Oncology Network. Results: From September 2017 to August 2021, 35 patients were enrolled. Of these, 29 (83%) had R0 resections, and 7 (20%) received adjuvant radiotherapy. Median age was 67 years, 21 (60.0%) female. Recurrence-free survival (RFS) rates at 1 and 2 years were 50% [95% confidence interval (CI), 31%–66%] and 37% (95% CI, 19%–55%), respectively. Overall survival rates at 1 and 2 years were 87% (95% CI, 68%–95%) and 68% (95% CI, 46%–83%), respectively. Median RFS was 10.3 months (95% CI, 5.7–25.8). Most common grade 3 toxicities were diarrhea (14%), hypertension (14%), and hyponatremia (11%), with no grade 4/5 toxicities. Conclusions: Flip-dose ipilimumab and nivolumab after resection of mucosal melanoma is associated with outcomes improved over that of surgical resection alone. Long-term follow-up, subgroup analyses and correlative studies are ongoing.
- Published
- 2023
6. Overall survival in the OlympiA phase III trial of adjuvant olaparib in patients with germline pathogenic variants in BRCA1/2 and high-risk, early breast cancer
- Author
-
C.E. Geyer, J.E. Garber, R.D. Gelber, G. Yothers, M. Taboada, L. Ross, P. Rastogi, K. Cui, A. Arahmani, G. Aktan, A.C. Armstrong, M. Arnedos, J. Balmaña, J. Bergh, J. Bliss, S. Delaloge, S.M. Domchek, A. Eisen, F. Elsafy, L.E. Fein, A. Fielding, J.M. Ford, S. Friedman, K.A. Gelmon, L. Gianni, M. Gnant, S.J. Hollingsworth, S.-A. Im, A. Jager, Ó. Þ Jóhannsson, S.R. Lakhani, W. Janni, B. Linderholm, T.-W. Liu, N. Loman, L. Korde, S. Loibl, P.C. Lucas, F. Marmé, E. Martinez de Dueñas, R. McConnell, K.-A. Phillips, M. Piccart, G. Rossi, R. Schmutzler, E. Senkus, Z. Shao, P. Sharma, C.F. Singer, T. Španić, E. Stickeler, M. Toi, T.A. Traina, G. Viale, G. Zoppoli, Y.H. Park, R. Yerushalmi, H. Yang, D. Pang, K.H. Jung, A. Mailliez, Z. Fan, I. Tennevet, J. Zhang, T. Nagy, G.S. Sonke, Q. Sun, M. Parton, M.A. Colleoni, M. Schmidt, A.M. Brufsky, W. Razaq, B. Kaufman, D. Cameron, C. Campbell, A.N.J. Tutt, Paul Sevelda, Ferdinand Haslbauer, Monika Penzinger, Leopold Öhler, Christoph Tinchon, Richard Greil, Sonja Heibl, Rupert Bartsch, Viktor Wette, Christian F. Singer, Claudia Pasterk, Ruth Helfgott, Gunda Pristauz-Telsnigg, Herbert Stöger, Angsar Weltermann, Daniel Egle, Irene Thiel, David Fuchs, Holger Rumpold, Kathrin Strasser-Weippl, Beate Rautenberg, Volkmar Müller, Marcus Schmidt, Stefan Paepke, Mustafa Aydogdu, Christoph Thomssen, Joachim Rom, Christine Mau, Peter Fasching, Uwe-Jochen Göhring, Thorsten Kühn, Stefanie Noeding, Sherko Kümmel, John Hackmann, Elmar Stickeler, Abhishek Joshi, Joanna Dewar, Michael Friedlander, Kelly-Anne Phillips, Yoland Antill, Natasha Woodward, Ehtesham Abdi, Susan Tiley, Mathew George, David Boadle, Annabel Goodwin, Andre van der Westhuizen, George Kannourakis, Nicholas Murray, Nicole McCarthy, Judith Kroep, Maaike de Boer, Joan Heijns, Agnes Jager, Franciscus Erdkamp, Sandra Bakker, Gabe S. Sonke, Amer Sami, John Mackey, Catherine Prady, Andrea Eisen, Christine Desbiens, Erica Patocskai, Cristiano Ferrario, Karen Gelmon, Louise Bordeleau, Haji Chalchal, Saroj Niraula, null ido wolf, Elżbieta Senkus, François Duhoux, null Randal d’Hondt, Sylvie Luce, Daphné t’Kint de Roodenbeke, Konstantinos Papadimitriou, Marleen Borms, Claire Quaghebeur, William Jacot, Etienne Brain, Laurence Venat-Bouvet, Alain Lortholary, Zbigniew Nowecki, Fátima Cardoso, Richard Hayward, Santiago Bella, Mauricio Fernández Lazzaro, Norma Pilnik, Luis E. Fein, Cesar Blajman, Guillermo Lerzo, Mirta Varela, Juan Jose Zarba, Diego Kaen, Maria Victoria Constanzo, Joke Tio, Wulf Siggelkow, Christian Jackisch, Eva Maria Grischke, Dirk Zahm, Sara Tato-Varela, Sabine Schmatloch, Peter Klare, Andrea Stefek, Kerstin Rhiem, Oliver Hoffmann, Mustafa Deryal, Isolde Gröll, Peter Ledwon, Christoph Uleer, Petra Krabisch, Jochem Potenberg, Maren Darsow, Tjoung-Won Park-Simon, Heinz-Gert Höffkes, Till-Oliver Emde, Gerd Graffunder, Oliver Tomé, Dirk Forstmeyer, Jürgen Terhaag, Christoph Salat, Karin Kast, Steffi Weniger, Carsten Schreiber, Bernhard Heinrich, Max Dieterich, Michaela Penelope Wüllner, Raquel Andrés Conejero, José Ángel García Sáenz, Lourdes Calvo Martinez, Angels Arcusa Lanza, Serafín Morales Murillo, Fernando Henao Carrasco, Salvador Blanch Tormo, Isabel Álvarez López, Juan Ignacio Delgado Mingorance, Elena Álvarez Gomez, Marta Santisteban, Josefina Cruz Jurado, Vanesa Quiroga, Manuel Ruiz Borrego, Eduardo Martínez de Dueñas, Jose Enrique Alés Martínez, Juan De la Haba, Noelia Martínez Jañez, Álvaro Rodríguez Lescure, Antonio Antón Torres, Gema Llort Crusades, Santiago González-Santiago, Antonia Marquez Aragones, Ana Laura Ortega, Agusti Barnadas Molins, José Ignacio Chacón López-Muñiz, Miguel Martín Jiménez, Ana Santaballa Bertrán, César Rodríguez, Lucía González Cortijo, Elisabetta Cretella, Laura Cortesi, Enzo Maria Ruggeri, Claudio Verusio, Stefania Gori, Andrea Bonetti, Anna Maria Mosconi, Oskar Johannsson, Guy Jerusalem, Patrick Neven, Tünde Nagy, Graziella Pinotti, Marco A. Colleoni, Antonio Bernardo, Lorenzo Gianni, Eraldo Bucci, Laura Biganzoli, Konstantin Dedes, Urban Novak, Khalil Zaman, Jeremy Braybrooke, Matthew Winter, Daniel Rea, Muireann Kelleher, Sophie Barrett, Stephen Chan, Tamas Hickish, Jane Hurwitz, John Conibear, Apurna Jegannathen, Marina Parton, Andrew Tutt, Rozenn Allerton, Annabel Borley, Anne Armstrong, Ellen Copson, Nicola Levitt, Jean Abraham, Timothy Perren, Rebecca Roylance, Kazushige Ishida, Tatsuya Toyama, Norikazu Masuda, Junichiro Watanabe, Eriko Tokunaga, Takayuki Kinoshita, Yoshiaki Rai, Masahiro Takada, Yasuhiro Yanagita, Rikiya Nakamura, Takahiro Nakayama, Yasuto Naoi, Hiroji Iwata, Seigo Nakamura, Masato Takahashi, Kenjiro Aogi, Koichiro Tsugawa, Hirofumi Mukai, Toshimi Takano, Akihiko Osaki, Nobuaki Sato, Hideko Yamauchi, Yutaka Tokuda, Mitsuya Ito, Takeki Sugimoto, Shakeela W. Bahadur, Patricia A. Ganz, Min J. Lu, Monica M. Mita, James Waisman, Jonathan A. Polikoff, Melinda L. Telli, Samantha A. Seaward, J. Marie Suga, Lara N. Durna, Jennifer Fu Carney, Alex Menter, Ajithkumar Puthillath, Nitin Rohatgi, James H. Feusner, Kristie A. Bobolis, Peter D. Eisenberg, Derrick Wong, Virginia F. Borges, Alexander T. Urquhart, Erin W. Hofstatter, Edward C. McCarron, Claudine Isaacs, Pia Herbolsheimer, Ramya Varadarajan, Adam Raben, Ruby Anne E. Deveras, Frances Valdes-Albini, Reshma L. Mahtani, Jane L. Meisel, Bradley T. Sumrall, Cheryl F. Jones, Samuel N. Ofori, Kenneth N.M. Sumida, Mark Karwal, Deborah W. Wilbur, (Joe) Singh, David M. Spector, John Schallenkamp, Douglas E. Merkel, Shelly S. Lo, Pam G. Khosla, Massimo Cristofanilli, Lisa Flaum, Kent F. Hoskins, Melody A. Cobleigh, Elyse A. Lambiase, Olwen M. Hahn, Ira A. Oliff, Bryan A. Faller, James L. Wade, Nafisa D. Burhani, Amaryllis Gil, Harvey E. Einhorn, Anna M.V. Storniolo, Brian K. Chang, Maitri Kalra, Erwin L. Robin, Bilal Ansari, Priyanka Sharma, Shaker R. Dakhil, Richard L. Deming, John T. Cole, David S. Hanson, Augusto C. Ochoa, Judy E. Garber, Harvey Zimbler, Deborah K. Armstrong, Katherine H.R. Tkaczuk, David A. Riseberg, Brian M. O'Connor, Thomas H. Openshaw, Dana Zakalik, Cynthia M. Vakhariya, Anne F. Schott, Michael S. Simon, Thomas J. Doyle, Tareq Al Baghdadi, Amy VanderWoude, Patrick J. Flynn, Richard T. Zera, Bret E.B. Friday, Kathryn J. Ruddy, Ron Smith, null Ademuyiwa, Foluso Olabisi, Robert Ellis, Jay W. Carlson, null Marchello, Benjamin T, Edward A. Levine, Paul K. Marcom, Cameron B. Harkness, Antoinette R. Tan, William J. Charles, Charles S. Kuzma, Shonda Asaad, James E. Radford, Preston D. Steen, Madhu Unnikrishnan, Grant R. Seeger, Kirsten M.H. Leu, Mehmet S. Copur, Ralph J. Hauke, Gamini S. Soori, Bradley A. Arrick, Jennifer G. Reeder, Deborah L. Toppmeyer, Zoneddy R. Dayao, Sylvia Adams, Eleni Andreopoulou, Magnuson Allison, Jesus D. Anampa Mesias, Ruby Sharma, Bhuvaneswari Ramaswamy, Aaron T. Gerds, Robert R. Shenk, Howard M. Gross, Shruti Trehan, Wajeeha Razaq, Abdul H. Mansoor, Christie J. Hilton, Adam M. Brufsky, Chanh Huynh, Nabila Chowdhury, Susan M. Domchek, Elin R. Sigurdson, Terrence P. Cescon, Marc A. Rovito, Albert S. DeNittis, Victor G. Vogel, Thomas B. Julian, L.E. Boyle, Luis Baez-Diaz, Frank J. Brescia, John E. Doster, Robert D. Siegel, Lucas Wong, Tejal Patel, Julie R. Nangia, Catherine A. Jones, George M. Cannon, Harry D. Bear, Hetal Vachhani, Mary Wilkinson, Marie E. Wood, Fengting Yan, Xingwei Sui, Carol M. van Haelst, Jennifer M. Specht, Ying Zhuo, Rubina Qamar, Matthew L. Ryan, Abigail Stockham, Shamsuddin Virani, Arlene A. Gayle, Steven J. Jubelirer, Sobha Kurian, Mohamad A. Salkeni, Niklas Loman, Barbro Linderholm, Gustav Silander, Anna-Lotta Hallbeck, Anna von Wachenfeldt Väppling, Elsa Curtit, Catarina Cardoso, Sofia Braga, Miguel Abreu, Mafalda Casa-Nova, Mónica Nave, Eva María Ciruelos Gil, Judith Balmaña Gelpi, Adela Fernández Ortega, Josep Gumà Padró, Begoña Bermejo de las Heras, María González Cao, Juan Cueva Bañuelos, Jesús Alarcon Company, Gemma Viñas Villaró, Laura García Estevez, Jens Huober, Steffi Busch, Tanja Fehm, Antje Hahn, Andrea Grafe, Thomas Noesselt, Thomas Dewitz, Harald Wagner, Christina Bechtner, Michael Weigel, Hans-Christian Kolberg, Thomas Decker, Jörg Thomalla, Tobias Hesse, Nadia Harbeck, Jan Schröder Jens-Uwe Blohmer, Marc Wolf Sütterlin, Renske Altena, Chang-Fang Chiu, Shin-Cheh Chen, Ming-Feng Hou, Yuan-Ching Chang, Shang-Hung Chen, Shou-Tung Chen, Chiun-Sheng Huang, Dah-Cherng Yeh, Jyh-Cherng Yu, Ling-Ming Tseng, Wei-Pang Chung, Audrey Mailliez, Thierry Petit, Suzette Delaloge, Christelle Lévy, Philippe Dalivoust, Jean-Marc Extra, Marie-Ange Mouret-Reynier, Anne-Claire Hardy-Bessard, Hélène Simon, Tiffenn L'Haridon, Alice Mege, Sylvie Giacchetti, Camille Chakiba-Brugere, Alain Gratet, Virginie Pottier, Jean-Marc Ferrero, Isabelle Tennevet, Christophe Perrin, Jean-Luc Canon, Sofie Joris, Zhimin Shao, Binghe Xu, ZeFei Jiang, Qiang Sun, Kunwei Shen, Da Pang, Jin Zhang, Shui Wang, Hongjian Yang, Ning Liao, Hong Zheng, Peifen Fu, Chuangui Song, Yongsheng Wang, Zhimin Fan, Cuizhi Geng, Olivier Tredan, László Landherr, Bella Kaufman, Rinat Yerushalmi, Beatrice Uziely, Pierfranco Conte, Claudio Zamagni, Giampaolo Bianchini, Michelino De Laurentiis, Carlo Tondini, Vittorio Gebbia, Mariangela Ciccarese, Tomasz Sarosiek, Jacek Mackiewicz, Anna Słowińska, Ewa Kalinka, Tomasz Huzarski, Seock-Ah Im, Kyung Hae Jung, Joo Hyuk Sohn, Jee Hyun Kim, Keun Seok Lee, Yeon Hee Park, Kyoung Eun Lee, Yee Soo Chae, Eun Kyung Cho, Institut Català de la Salut, [Geyer CE Jr] NRG Oncology/NSABP Foundation, Pittsburgh, USA. Department of Medicine, UPMC Hillman Cancer Center, Pittsburgh, USA. [Garber JE] Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA. [Gelber RD] Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA. Harvard T.H. Chan School of Public Health, Boston, USA. Frontier Science Foundation, Boston, USA. [Yothers G] NRG Oncology/NSABP Foundation, Pittsburgh, USA. Department of Biostatistics, University of Pittsburgh, Pittsburgh, USA. [Taboada M] Oncology Biometrics Department, AstraZeneca, Macclesfield, UK. [Ross L] Department of Data Management, Frontier Science (Scotland), Kincraig, Scotland, UK. [Balmaña J] Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. Servei d’Oncologia Mèdica, Vall d’Hebron Hospital Universitari, Barcelona, Spain, Vall d'Hebron Barcelona Hospital Campus, Medical Oncology, Public Health, Virology, Department of Psychology, Education and Child Studies, Internal Medicine, General Practice, and Child and Adolescent Psychiatry / Psychology
- Subjects
Neoplasms::Neoplasms by Site::Breast Neoplasms [DISEASES] ,Medicaments antineoplàstics - Ús terapèutic ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,Breast Neoplasms ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,olaparib ,Article ,breast cancer ,SDG 3 - Good Health and Well-being ,BRCA1/2 ,células::células germinativas [ANATOMÍA] ,Humans ,Other subheadings::/therapeutic use [Other subheadings] ,Cells::Germ Cells [ANATOMY] ,neoplasias::neoplasias por localización::neoplasias de la mama [ENFERMEDADES] ,Otros calificadores::/uso terapéutico [Otros calificadores] ,BRCA1 Protein ,PARP inhibition ,acciones y usos químicos::acciones farmacológicas::usos terapéuticos::antineoplásicos [COMPUESTOS QUÍMICOS Y DROGAS] ,adjuvant therapy ,Hematology ,Cèl·lules germinals ,Germ Cells ,Oncology ,Mama - Càncer - Tractament ,Phthalazines ,Female ,Chemical Actions and Uses::Pharmacologic Actions::Therapeutic Uses::Antineoplastic Agents [CHEMICALS AND DRUGS] - Abstract
Adjuvant therapy; Breast cancer; Olaparib Terapia adyuvante; Cáncer de mama; Olaparib Teràpia adjuvant; Càncer de mama; Olaparib Background The randomized, double-blind OlympiA trial compared 1 year of the oral poly(adenosine diphosphate-ribose) polymerase inhibitor, olaparib, to matching placebo as adjuvant therapy for patients with pathogenic or likely pathogenic variants in germline BRCA1 or BRCA2 (gBRCA1/2pv) and high-risk, human epidermal growth factor receptor 2-negative, early breast cancer (EBC). The first pre-specified interim analysis (IA) previously demonstrated statistically significant improvement in invasive disease-free survival (IDFS) and distant disease-free survival (DDFS). The olaparib group had fewer deaths than the placebo group, but the difference did not reach statistical significance for overall survival (OS). We now report the pre-specified second IA of OS with updates of IDFS, DDFS, and safety. Patients and methods One thousand eight hundred and thirty-six patients were randomly assigned to olaparib or placebo following (neo)adjuvant chemotherapy, surgery, and radiation therapy if indicated. Endocrine therapy was given concurrently with study medication for hormone receptor-positive cancers. Statistical significance for OS at this IA required P < 0.015. Results With a median follow-up of 3.5 years, the second IA of OS demonstrated significant improvement in the olaparib group relative to the placebo group [hazard ratio 0.68; 98.5% confidence interval (CI) 0.47-0.97; P = 0.009]. Four-year OS was 89.8% in the olaparib group and 86.4% in the placebo group (Δ 3.4%, 95% CI −0.1% to 6.8%). Four-year IDFS for the olaparib group versus placebo group was 82.7% versus 75.4% (Δ 7.3%, 95% CI 3.0% to 11.5%) and 4-year DDFS was 86.5% versus 79.1% (Δ 7.4%, 95% CI 3.6% to 11.3%), respectively. Subset analyses for OS, IDFS, and DDFS demonstrated benefit across major subgroups. No new safety signals were identified including no new cases of acute myeloid leukemia or myelodysplastic syndrome. Conclusion With 3.5 years of median follow-up, OlympiA demonstrates statistically significant improvement in OS with adjuvant olaparib compared with placebo for gBRCA1/2pv-associated EBC and maintained improvements in the previously reported, statistically significant endpoints of IDFS and DDFS with no new safety signals. Funding for this work, which was conducted as a collaborative partnership among the Breast International Group, NRG Oncology, Frontier Science, AstraZeneca, and Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, U.S.A. (MSD), was provided by the National Institutes of Health (grant numbers: U10CA 180868, UG1CA 189867, and U10CA 180822) and by AstraZeneca as part of an alliance between AstraZeneca and MSD. Provision of olaparib and placebo was from AstraZeneca.
- Published
- 2022
7. Data from SALVO: Single-Arm Trial of Ipilimumab and Nivolumab as Adjuvant Therapy for Resected Mucosal Melanoma
- Author
-
Robert R. McWilliams, Tina Hieken, Matthew Stephen Block, Mohammed M. Milhem, Sarah Schober, Brendan D. Curti, Ralph J. Hauke, Evidio Domingo-Musibay, Yousef Zakharia, Anthony J. Olszanski, Gregory Russell Pond, and Lisa A. Kottschade
- Abstract
Purpose:Mucosal melanoma is a rare, aggressive form of melanoma with extremely high recurrence rates despite definitive surgical resection with curative intent. Currently there is no consensus on adjuvant therapy. Data on checkpoint inhibitors for adjuvant therapy are lacking.Patients and Methods:We performed a single-arm, multicenter clinical trial using “flip dose” ipilimumab (1 mg/kg q3w × 4 cycles), and nivolumab (3 mg/kg q3w × 4 cycles), then nivolumab 480 mg q4w × 11 cycles to complete a year of adjuvant therapy. Participants must have had R0/R1 resection ≤90 days before registration, no prior systemic therapy (adjuvant radiotherapy allowed), ECOG 0/1, and no uncontrolled autoimmune disease or other invasive cancer. Patients were recruited through the Midwest Melanoma Partnership/Hoosier Oncology Network.Results:From September 2017 to August 2021, 35 patients were enrolled. Of these, 29 (83%) had R0 resections, and 7 (20%) received adjuvant radiotherapy. Median age was 67 years, 21 (60.0%) female. Recurrence-free survival (RFS) rates at 1 and 2 years were 50% [95% confidence interval (CI), 31%–66%] and 37% (95% CI, 19%–55%), respectively. Overall survival rates at 1 and 2 years were 87% (95% CI, 68%–95%) and 68% (95% CI, 46%–83%), respectively. Median RFS was 10.3 months (95% CI, 5.7–25.8). Most common grade 3 toxicities were diarrhea (14%), hypertension (14%), and hyponatremia (11%), with no grade 4/5 toxicities.Conclusions:Flip-dose ipilimumab and nivolumab after resection of mucosal melanoma is associated with outcomes improved over that of surgical resection alone. Long-term follow-up, subgroup analyses and correlative studies are ongoing.
- Published
- 2023
8. Supplementary Table S2 from SALVO: Single-Arm Trial of Ipilimumab and Nivolumab as Adjuvant Therapy for Resected Mucosal Melanoma
- Author
-
Robert R. McWilliams, Tina Hieken, Matthew Stephen Block, Mohammed M. Milhem, Sarah Schober, Brendan D. Curti, Ralph J. Hauke, Evidio Domingo-Musibay, Yousef Zakharia, Anthony J. Olszanski, Gregory Russell Pond, and Lisa A. Kottschade
- Abstract
Grade 3+ Adverse Events
- Published
- 2023
9. Supplementary Figure 1 from Safety and Antitumor Activity of Apalutamide (ARN-509) in Metastatic Castration-Resistant Prostate Cancer with and without Prior Abiraterone Acetate and Prednisone
- Author
-
Howard I. Scher, Margaret K. Yu, Carla J. de Boer, Edna Chow Maneval, Rajesh Bandekar, Ralph J. Hauke, Mansoor Saleh, Charles J. Ryan, Joshi J. Alumkal, Ronald F. Tutrone, Neal D. Shore, Emmanuel S. Antonarakis, and Dana E. Rathkopf
- Abstract
Study design (A) and patient disposition (B); data cutoff December 31, 2014.
- Published
- 2023
10. Data from Safety and Antitumor Activity of Apalutamide (ARN-509) in Metastatic Castration-Resistant Prostate Cancer with and without Prior Abiraterone Acetate and Prednisone
- Author
-
Howard I. Scher, Margaret K. Yu, Carla J. de Boer, Edna Chow Maneval, Rajesh Bandekar, Ralph J. Hauke, Mansoor Saleh, Charles J. Ryan, Joshi J. Alumkal, Ronald F. Tutrone, Neal D. Shore, Emmanuel S. Antonarakis, and Dana E. Rathkopf
- Abstract
Purpose: To evaluate the efficacy of apalutamide before or after treatment with abiraterone acetate and prednisone (AAP) in patients with progressive metastatic castration-resistant prostate cancer (mCRPC).Experimental Design: Two cohorts were studied: AAP-naïve and post-AAP patients who had received ≥6 months of AAP. Patients had progressive mCRPC per rising prostate-specific antigen (PSA) and/or imaging, without prior chemotherapy exposure. All received apalutamide 240 mg/day. Primary endpoint was ≥50% decline in 12-week PSA according to Prostate Cancer Working Group 2 criteria. Secondary endpoints included time to PSA progression and time on treatment.Results: Forty-six patients enrolled in the AAP-naïve (n = 25) and post-AAP (n = 21) cohorts. The 12-week PSA response rate was 88% (22/25) and 22% (4/18), median time to PSA progression was 18.2 months [95% confidence interval (CI), 8.3 months–not reached) and 3.7 months (95% CI, 2.8–5.6 months), and median time on treatment 21 months (range, 2.6–37.5) and 4.9 months (range, 1.3–23.2), for the AAP-naïve and post-AAP cohorts, respectively. Eighty percent (95% CI, 59–93) and 64% (95% CI, 43–82) of AAP-naïve and 43% (95% CI, 22–66) and 10% (95% CI, 1–30) of post-AAP patients remained on treatment for 6+ and 12+ months, respectively. Common treatment-emergent adverse events in both cohorts were grade 1 or 2 fatigue, diarrhea, nausea, and abdominal pain.Conclusions: Apalutamide was safe, well tolerated, and demonstrated clinical activity in mCRPC, with 80% of AAP-naïve and 43% of post-AAP patients, remaining on treatment for 6 months or longer. Clin Cancer Res; 23(14); 3544–51. ©2017 AACR.
- Published
- 2023
11. Sequential Therapy in Metastatic Renal Cell Carcinoma
- Author
-
Bradford R Hirsch, John M Burke, Manish Agrawal, Ralph J Hauke, Thomas E Hutson, Gury Doshi, Mark T Fleming, and Nicholas J Vogelzang
- Subjects
sequential therapy ,metastasis ,renal cell carcinoma ,Diseases of the genitourinary system. Urology ,RC870-923 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The treatment of metastatic renal cell carcinoma (mRCC) has changed dramatically in the past decade. As the number of available agents, and related volume of research, has grown, it is increasingly complex to know how to optimally treat patients. The authors are practicing medical oncologists at the US Oncology Network, the largest community-based network of oncology providers in the country, and represent the leadership of the Network's Genitourinary Research Committee. We outline our thought process in approaching sequential therapy of mRCC and the use of real-world data to inform our approach. We also highlight the evolving literature that will impact practicing oncologists in the near future.
- Published
- 2016
- Full Text
- View/download PDF
12. Chemotherapy options in castration-resistant prostate cancer
- Author
-
Benjamin A Teply and Ralph J Hauke
- Subjects
Biomarkers ,chemotherapy ,prostate cancer ,Diseases of the genitourinary system. Urology ,RC870-923 - Abstract
Introduction: The treatment landscape for patients with metastatic castration-resistant prostate cancer (CRPC) is evolving, with recent approvals of immune therapy, novel hormonal therapy, and bone-targeted therapy. Chemotherapy remains an essential component of the armamentarium. Herein, we review current chemotherapy options for patients with CRPC and discuss future challenges. Methods: We reviewed literature for chemotherapy agents in prostate cancer, with special attention to the evidence for efficacy of the currently approved agents. We also reviewed emerging data on biomarkers of response to chemotherapy for CRPC. Results: Taxanes, especially docetaxel and cabazitaxel, have first- and second-line indications for CRPC, respectively, with both providing a survival benefit. Multiple attempts to improve on the single agent efficacy of docetaxel with combination therapy have not generally been successful although platinum combinations are used for resistant phenotypes. Reductions in prostate-specific antigen by ≥30% and reductions in circulating tumor cells (CTCs) to ≤ 5 are associated with improved survival on chemotherapy. Chemotherapy may continue to be effective therapy for patients with biomarkers that are associated with resistance to androgen-directed therapies (androgen receptor splice variant 7 positivity in CTCs or high CTC heterogeneity). Conclusions: Chemotherapy remains an essential component of CRPC therapy, and biomarkers are being identified to define clinical scenarios where chemotherapy may be the optimal therapy choice.
- Published
- 2016
- Full Text
- View/download PDF
13. Management of Metastatic Clear Cell Renal Cell Carcinoma: ASCO Guideline
- Author
-
W. Kimryn Rathmell, R. Bryan Rumble, Peter J. Van Veldhuizen, Hikmat Al-Ahmadie, Hamid Emamekhoo, Ralph J. Hauke, Alexander V. Louie, Matthew I. Milowsky, Ana M. Molina, Tracy L. Rose, Shankar Siva, Nicholas G. Zaorsky, Tian Zhang, Rubina Qamar, Terry M. Kungel, Bryan Lewis, and Eric A. Singer
- Subjects
Vascular Endothelial Growth Factor A ,Cancer Research ,Oncology ,Humans ,Angiogenesis Inhibitors ,Carcinoma, Renal Cell ,Protein Kinase Inhibitors ,Kidney Neoplasms - Abstract
PURPOSE To provide recommendations for the management of patients with metastatic clear cell renal cell carcinoma (ccRCC). METHODS An Expert Panel conducted a systematic literature review to obtain evidence to guide treatment recommendations. RESULTS The panel considered peer-reviewed reports published in English. RECOMMENDATIONS The diagnosis of metastatic ccRCC should be made using tissue biopsy of the primary tumor or a metastatic site with the inclusion of markers and/or stains to support the diagnosis. The International Metastatic RCC Database Consortium risk criteria should be used to inform treatment. Cytoreductive nephrectomy may be offered to select patients with kidney-in-place and favorable- or intermediate-risk disease. For those who have already had a nephrectomy, an initial period of active surveillance may be offered if they are asymptomatic with a low burden of disease. Patients with favorable-risk disease who need systemic therapy may be offered an immune checkpoint inhibitor (ICI) in combination with a vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitor (TKI); patients with intermediate or poor risk should be offered a doublet regimen (no recommendation was provided between ICIs or an ICI in combination with a VEGFR TKI). For select patients, monotherapy with either an ICI or a VEGFR TKI may be offered on the basis of comorbidities. Interleukin-2 remains an option, although selection criteria could not be identified. Recommendations are also provided for second- and subsequent-line therapy as well as the treatment of bone metastases, brain metastases, or the presence of sarcomatoid features. Participation in clinical trials is highly encouraged for patients with metastatic ccRCC. Additional information is available at www.asco.org/genitourinary-cancer-guidelines
- Published
- 2022
14. Incorporating radioligand therapy in clinical practice in the United States for patients with prostate cancer
- Author
-
Jeremie Calais, Stephen M. Eulau, Linda Gardner, Ralph J. Hauke, Ayse T. Kendi, Neal D. Shore, and Song Zhao
- Subjects
Oncology ,Radiology, Nuclear Medicine and imaging ,General Medicine - Published
- 2023
15. 389 Phase II of CD40 agonistic antibody sotigalimab (APX005M) in combination with nivolumab in subjects with metastatic melanoma with confirmed disease progression on anti-PD-1 therapy
- Author
-
Montaser Shaheen, Lynn M. Schuchter, Delvys Rodriguez-Abreu, Erin L. Filbert, Maria Gonzalez-Cao, Apar Kishor Ganti, Ralph J. Hauke, Enriqueta Felip, Nicholas Iannotti, Ana Arance, Sarah A. Weiss, Harriet M. Kluger, Valentina Boni, Mario Sznol, Alfonso Berrocal, Miguel-Ángel Berciano-Guerrero, and Gerald P. Linette
- Subjects
Pharmacology ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Nausea ,Melanoma ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Rash ,Tolerability ,Internal medicine ,Molecular Medicine ,Immunology and Allergy ,Medicine ,Chills ,medicine.symptom ,Nivolumab ,business ,Off Treatment ,Progressive disease ,RC254-282 - Abstract
BackgroundA significant number of melanoma patients treated with anti-PD-1 alone or in combination with anti-CTLA-4 have transient or no response to treatment. Sotigalimab is a CD40 agonist antibody with unique epitope specificity and Fc receptor binding profile for optimal therapeutic application. Preclinical studies suggest that sotigalimab can be combined with PD-1 blockade to trigger effective anti-tumor immunity. We conducted a multi-center, open label, Phase Ib-parallel arm phase II trial (NCT03123783) to evaluate the combination of sotigalimab with nivolumab in subjects with anti-PD-1/PD-L1 refractory metastatic melanoma.MethodsThe study objective was to evaluate the efficacy and safety of sotigalimab in combination with nivolumab in anti-PD-1/PD-L1 refractory advanced melanoma patients. Subjects received sotigalimab (0.3mg/kg) combined with nivolumab (360mg) every 3 weeks. Thirty-eight subjects with unresectable or metastatic melanoma who had confirmed progressive disease during treatment with anti-PD-1 therapy (documented by 2 consecutive tumor assessments) were enrolled (evaluable for safety) and 33 subjects were evaluable for efficacy.ResultsSix subjects had PR (including one unconfirmed PR) for an ORR of 18%. The mDOR was 18.7 months. Two subjects with PR received treatment for >2 years. Three of the six responding subjects remain off all therapy for ≥26 months, and one patient required stereotactic radiosurgery to a single brain lesion ten months after stopping therapy and has not required additional local or systemic therapy since. Three additional subjects had prolonged SD (12.6, 7.6, 6.2 months). The DCR was 48% and 33% of subjects experienced reduction in target lesions. Efficacy was observed in patients regardless of their tumor PD-L1 expression. The overall safety profile of the combination is consistent with the profiles of individual agents. The majority of AEs observed were of mild to moderate intensity (CTCAE Grade ≤2). The most commonly observed AEs were: pyrexia, chills, nausea, fatigue, pruritus, transaminitis, headache, asthenia, myalgia, rash, vomiting and arthralgia. There were no Grade 4 or 5 AEs related to study drugs. There were no treatment discontinuations due to AEs.ConclusionsThe combination of sotigalimab and nivolumab demonstrated treatment benefit (tumor response or prolonged disease control) in anti-PD-1/PD-L1 refractory melanoma patients with an overall favorable safety and tolerability profile. Notably, a subset of patients remain in response off treatment for ≥26 months. These results warrant further study of this combination in advanced, refractory melanoma.AcknowledgementsWe extend our gratitude to the patients and their families who made this trial possible and the clinical study teams involved in this trial. We thank BMS for providing the nivolumab for this study.Trial RegistrationNCT03123783Ethics ApprovalThis study was approved by the Institutional Review Boards at Yale University (#20170300), University of Nebraska Medical Center (#543-18-CB) and The Hospital Regional de Málaga (#19.03.1341E1-GHM).
- Published
- 2021
16. SALVO: Single-arm trial of ipilimumab and nivolumab as adjuvant therapy for resected mucosal melanoma
- Author
-
Lisa A. Kottschade, Gregory Russell Pond, Anthony J. Olszanski, Yousef Zakharia, Evidio Domingo-Musibay, Ralph J. Hauke, Brendan D. Curti, Sarah Schober, Mohammed M. Milhem, Matthew Stephen Block, and Robert R. McWilliams
- Subjects
Cancer Research ,Oncology - Abstract
9573 Background: Mucosal melanoma is a rare, highly aggressive form of melanoma with extremely high recurrence rates, despite definitive surgical resection. Median RFS has been reported to be 5.4m, with RFS rates at 1 and 2 years of 10%, and 0%, respectively (Lian B, Si L, Cui C, et al. Phase II Randomized Trial Comparing High-Dose IFN-α2b with Temozolomide Plus Cisplatin as Systemic Adjuvant Therapy for Resected Mucosal Melanoma. Clinical Cancer Research 2013, 19(16):4488-4498). Currently there is no consensus on recommendations for adjuvant therapy. Data on the use of immune checkpoint inhibitors (ICI) adjuvantly is lacking. Methods: We performed a single arm, multicenter clinical trial using “flip dose” ipilimumab (1mg/kg q3w x4 cycles),and nivolumab (3 mg.kg q3w x4 cycles), then Nivolumab 480 mg q4w x 11 cycles to complete a year of adjuvant therapy. The primary endpoint was recurrence-free survival (RFS), and the study had 85% power to detect an improvement in RFS between 5.5 and 9.5 months using a one-sided log rank test. Participants must have had R0/R1 resection
- Published
- 2022
17. Cabozantinib (C) in combination with atezolizumab (A) in urothelial carcinoma (UC): Results from Cohorts 3, 4, 5 of the COSMIC-021 study
- Author
-
Sumanta K. Pal, Neeraj Agarwal, Parminder Singh, Andrea Necchi, Bradley Alexander McGregor, Ralph J. Hauke, Thomas Powles, Cristina Suárez, Carla M.L.- Van Herpen, Ulka N. Vaishampayan, Ramu Sudhagoni, Dominic Curran, Lana Andrianova, and Yohann Loriot
- Subjects
Cancer Research ,Oncology - Abstract
4504 Background: C, a multitargeted receptor tyrosine kinase inhibitor (TKI), promotes an immune-permissive environment that may enhance response to immune checkpoint inhibitors (ICIs). COSMIC-021, a multicenter phase 1b study, is evaluating C + A (anti‒PD-L1 therapy) in various solid tumors (NCT03170960). C + A demonstrated encouraging clinical activity in cohort 2 of COSMIC-021 in patients (pts) with UC previously treated with platinum-containing chemotherapy (chemo) (Pal S et al. ASCO 2020. Abstract 5013). Outcomes of C + A from 3 other UC cohorts (C3, C4, C5) are presented. Methods: Pts with inoperable locally advanced/metastatic UC with transitional cell histology and ECOG PS 0‒1 were eligible. Pts enrolled in C3 and C4 had no prior therapy and were cisplatin-based chemo ineligible (C3) or eligible (C4). C5 enrolled pts with one prior ICI and no prior VEGFR-TKI therapy. Pts received C 40 mg PO QD and A 1200 mg IV Q3W. CT/MRI scans were performed Q6W for first year and Q12W thereafter. The primary endpoint is objective response rate (ORR) per RECIST v1.1 by investigator. Other endpoints: safety, duration of response (DOR), PFS, and OS. Results: Thirty pts each were enrolled in C3 and C4, and 31 in C5. Baseline characteristics for C3, C4, and C5, respectively: median age, 74 y, 66 y, 68 y; male, 67%, 73%, 55%; ECOG PS 1, 63%, 57%, 74%; lung/liver metastasis; 33%/17%, 40%/20%, 58%/23%; ≥3 tumor sites, 30%, 43%, 45%; bladder as primary site, 67%, 70%, 71%. As of Nov 30, 2021, the median follow-up for C3, C4, and C5 was 27.9, 19.1, and 32.9 mo, respectively, with 1, 6, and 1 pts on treatment. C + A demonstrated clinical benefit across all cohorts (Table). Most common treatment-related adverse events (TRAEs) of any grade across C3, C4, and C5, respectively, were diarrhea (43%, 33%, 35%), nausea (27%, 17%, 26%), fatigue (27%, 27%, 48%), and decreased appetite (33%, 27%, 39%); grade 3/4 TRAEs occurred in 63%, 43%, and 45%, and there was no grade 5 TRAE. Conclusions: C + A demonstrated encouraging clinical activity with manageable toxicity in inoperable locally advanced/metastatic UC as first-line systemic therapy in cisplatin-based chemo eligible/ineligible pts and as second- or later line in pts who received prior ICI. Clinical trial information: NCT03170960. [Table: see text]
- Published
- 2022
18. TRANSFORMER: A Randomized Phase II Study Comparing Bipolar Androgen Therapy Versus Enzalutamide in Asymptomatic Men With Castration-Resistant Metastatic Prostate Cancer
- Author
-
Michael T. Schweizer, Mario A. Eisenberger, Jeffrey M. Holzbeierlein, Vanessa Bolejack, Przemyslaw Twardowski, Su Jin Lim, Ting Wang, Michael A. Carducci, Hao Wang, Russell Z. Szmulewitz, Oliver Sartor, Guru Sonpavde, Neeraj Agarwal, Thomas W. Flaig, Arif Hussain, Mark C. Markowski, Harry Cao, Rehab Abdallah, Wei Fu, Emmanuel S. Antonarakis, Ralph J. Hauke, Mark N. Stein, Shifeng Mao, Samuel R. Denmeade, Channing J. Paller, Jorge A. Garcia, Vasileios J. Assikis, and David Smith
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,medicine.drug_class ,Phases of clinical research ,urologic and male genital diseases ,Asymptomatic ,03 medical and health sciences ,Prostate cancer ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Nitriles ,Phenylthiohydantoin ,medicine ,Enzalutamide ,Humans ,Testosterone ,Neoplasm Metastasis ,Aged ,Aged, 80 and over ,Cross-Over Studies ,business.industry ,Cancer ,ORIGINAL REPORTS ,Middle Aged ,Prostate-Specific Antigen ,medicine.disease ,Androgen ,Androgen receptor ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,chemistry ,Androgen Therapy ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Asymptomatic Diseases ,Benzamides ,Quality of Life ,medicine.symptom ,business - Abstract
PURPOSE Prostate cancer (PCa) becomes resistant to androgen ablation through adaptive upregulation of the androgen receptor in response to the low-testosterone microenvironment. Bipolar androgen therapy (BAT), defined as rapid cycling between high and low serum testosterone, disrupts this adaptive regulation in castration-resistant PCa (CRPC). METHODS The TRANSFORMER (Testosterone Revival Abolishes Negative Symptoms, Fosters Objective Response and Modulates Enzalutamide Resistance) study is a randomized study comparing monthly BAT (n = 94) with enzalutamide (n = 101). The primary end point was clinical or radiographic progression-free survival (PFS); crossover was permitted at progression. Secondary end points included overall survival (OS), prostate-specific antigen (PSA) and objective response rates, PFS from randomization through crossover (PFS2), safety, and quality of life (QoL). RESULTS The PFS was 5.7 months for both arms (hazard ratio [HR], 1.14; 95% CI, 0.83 to 1.55; P = .42). For BAT, 50% decline in PSA (PSA50) was 28.2% of patients versus 25.3% for enzalutamide. At crossover, PSA50 response occurred in 77.8% of patients crossing to enzalutamide and 23.4% to BAT. The PSA-PFS for enzalutamide increased from 3.8 months after abiraterone to 10.9 months after BAT. The PFS2 for BAT→enzalutamide was 28.2 versus 19.6 months for enzalutamide→BAT (HR, 0.44; 95% CI, 0.22 to 0.88; P = .02). OS was 32.9 months for BAT versus 29.0 months for enzalutamide (HR, 0.95; 95% CI, 0.66 to 1.39; P = .80). OS was 37.1 months for patients crossing from BAT to enzalutamide versus 30.2 months for the opposite sequence (HR, 0.68; 95% CI, 0.36 to 1.28; P = .225). BAT adverse events were primarily grade 1-2. Patient-reported QoL consistently favored BAT. CONCLUSION This randomized trial establishes meaningful clinical activity and safety of BAT and supports additional study to determine its optimal clinical integration. BAT can sensitize CRPC to subsequent antiandrogen therapy. Further study is required to confirm whether sequential therapy with BAT and enzalutamide can improve survival in men with CRPC.
- Published
- 2021
19. Long-term progression-free survival of patients with metastatic melanoma or renal cell carcinoma following high-dose interleukin-2
- Author
-
Ralph J. Hauke, Richard L. White, Douglas B. Johnson, Ajjai Alva, Theodore F. Logan, Elizabeth J. Davis, Peter H. Wiernik, Gerald P Miletello, Asim Amin, Howard L. Kaufman, Brendan Curti, Joseph I. Clark, Ulka N. Vaishampayan, and Janice P. Dutcher
- Subjects
Oncology ,Interleukin 2 ,medicine.medical_specialty ,Metastatic melanoma ,medicine.medical_treatment ,Improved survival ,Systemic therapy ,adoptive ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Renal cell carcinoma ,Internal medicine ,medicine ,In patient ,Progression-free survival ,business.industry ,Brief Report ,General Medicine ,Immunotherapy ,medicine.disease ,cytokines ,030220 oncology & carcinogenesis ,immunotherapy ,business ,030215 immunology ,medicine.drug - Abstract
High-dose interleukin-2 (HD IL-2) was approved in the 1990s after demonstrating durable complete responses (CRs) in some patients with metastatic melanoma (mM) and metastatic renal cell carcinoma (mRCC). Patients who achieve this level of disease control have also demonstrated improved survival compared with patients who progress, but limited data are available describing the long-term course. The aim of this study was to better characterize long-term survival following successful HD IL-2 treatment in patients with no subsequent systemic therapy. Eleven HD IL-2 treatment centers identified patients with survival ≥5 years after HD IL-2, with no subsequent systemic therapy. Survival was evaluated from the date of IL-2 treatment to June 2017. Treatment courses consisted of 2 1-week cycles of HD IL-2. Patients were treated with HD IL-2 alone, or HD IL-2 followed by local therapy to achieve maximal response. 100 patients are reported: 54 patients with mM and 46 patients with mRCC. Progression-free survival (PFS) after HD IL-2 ranges from 5+ years to 30+ years, with a median follow-up of 10+ years. 27 mRCC and 32 mM are alive ≥10 years after IL-2. Thus, a small subset of patients with mM and mRCC achieve long-term PFS (≥5 years) after treatment with HD IL-2 as their only systemic therapy. The ability of HD IL-2 therapy to induce prolonged PFS should be a major consideration in studies of new immunotherapy combinations for mM and mRCC.
- Published
- 2021
20. An Open–Label, Randomized, Multi–Center Study Comparing the Sequence of High Dose Aldesleukin (Interleukin–2) and Ipilimumab (Yervoy) in Patients with Metastatic Melanoma
- Author
-
Bret Taback, William H. Sharfman, Merve Hasanov, Gary C. Doolittle, Lee D. Cranmer, Lawrence E. Flaherty, Sigrun Hallmeyer, Nancy C. Gregory, Ralph J. Hauke, Gregory A. Daniels, Lynn G. Feun, Sapna Pradyuman Patel, Denái R. Milton, and Mohammed M. Milhem
- Subjects
Interleukin 2 ,medicine.medical_specialty ,Metastatic melanoma ,Immunology ,Population ,Ipilimumab ,Gastroenterology ,Aldesleukin ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Immunology and Allergy ,ipilimumab ,Adverse effect ,education ,Melanoma ,RC254-282 ,education.field_of_study ,high dose interleukin-2 ,Intention-to-treat analysis ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,clinical trial ,RC581-607 ,Recombinant Proteins ,Clinical trial ,Nivolumab ,Oncology ,Interleukin-2 ,Immunologic diseases. Allergy ,business ,Research Article ,medicine.drug ,metastatic melanoma - Abstract
Combination immunotherapy with sequential administration may enhance metastatic melanoma (MM) patients with long-term disease control. High Dose Aldesleukin/Recombinant Interleukin-2 (HD rIL-2) and ipilimumab (IPI) offer complementary mechanisms against MM. This phase IV study assessed the sequenced use of HD rIL-2 and IPI in MM patients. Eligible Stage IV MM patients were randomized to treatment with either two courses of HD rIL-2(600,000 IU/kg) followed by four doses of IPI 3 mg/kg or vice-versa. The primary objective was to compare one-year overall survival (OS) with historical control (46%, Hodi et al., NEJM 2010). Secondary objectives were 1-year progression-free survival (PFS), objective response rate (ORR), and adverse events (AEs) profile. Evaluable Population (EP) included patients who received at least 50% of planned treatment with each drug. Thirteen and 16 patients were randomized to receive HD rIL-2 first, and IPI first, respectively. One-year OS rate was 75% for intention to treat population. Eighteen patients were included in EP, 8 in HD rIL-2, 10 in IPI first arm. In EP, 1-year OS, PFS and ORR rates were 87%, 68%, and 50%, respectively. The frequency of AEs was similar in both arms with 13 patients experiencing Grade 3 or higher AEs, 3 resulting in the end of study participation. There was one HD rIL-2-related death, from cerebral hemorrhage due to thrombocytopenia. In this study with small sample size, HD rIL-2 and IPI were safe to administer sequentially in MM patients and showed more than additive effects. 1-year OS was superior to that of IPI alone from historical studies.
- Published
- 2021
21. 405 CDX1140–01, a phase 1 dose-escalation/expansion study of CDX-1140 alone (Part 1) and in combination with CDX-301 (Part 2) or pembrolizumab (Part 3)
- Author
-
Danny N. Khalil, Tracey Rawls, Tibor Keler, Rachel E. Sanborn, Diego Alvarado, Lawrence J. Thomas, Laura Vitale, Maen Abdelrahim, Michael J. Yellin, Thomas U. Marron, Thomas Hawthorne, Nina Bhardwaj, Ralph J. Hauke, Mark H. O'Hara, Rodolfo Bordoni, Mark Rogalski, Nashat Y. Gabrail, and Michael S. Gordon
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Chemotherapy ,business.industry ,Nausea ,medicine.medical_treatment ,Follicular lymphoma ,Pembrolizumab ,medicine.disease ,Gastroenterology ,03 medical and health sciences ,Cytokine release syndrome ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,Vomiting ,Medicine ,Chills ,medicine.symptom ,business ,Pneumonitis - Abstract
Background CDX-1140 is an agonist anti-CD40 mAb selected to optimize systemic exposure and hence tumor microenvironment (TME) ingress. CDX-1140 activity may be enhanced by combining with CDX-301 (recombinant Flt3L), a dendritic cell growth factor, or with pembrolizumab, an anti-PD-1 mAb. Methods Patients with advanced solid or hematologic (Part 1 only) tumors are enrolled. Part 1 dose-escalation results have been presented (SITC 2019). In Part 2, CDX-1140 dose-escalation (0.09–1.5 mg/kg q4w) is in combination with CDX-301 (75 mcg/kg sc QD x 5 for 2 cycles). In Part 3, CDX-1140 dose-escalation (0.72–1.5 mg/kg q3w) is in combination with pembrolizumab 200 mg q3w. Part 1 and 2 expansion cohorts are dosed at the CDX-1140 MTD, 1.5 mg/kg q4w. Part 3 expansion cohorts are planned. Peripheral blood and tumor biomarkers analysis are ongoing. Results 92 patients have been treated (Part 1 n=57, Part 2 n=31, Part 3 n=4). Part 1 expansion cohorts in SCCHN (n=7) and RCC (n=5) are fully enrolled. Part 2 dose-escalation completed to the highest CDX-1140 dose and a SCCHN expansion cohort is ongoing. Part 3 dose-escalation recently initiated. Safety data is available for 23 and 10 patients at the MTD in Part 1 and 2, respectively. In general, the safety profiles were similar, with arthralgia (52% vs. 50%), pyrexia (44% vs 50%), fatigue (30% vs. 50%), chills (39% vs. 40%), vomiting (30% vs. 20%), nausea (26% vs 40%), myalgia (22% vs. 30%), increased ALT (22% vs. 20%), and increased AST (22% vs. 30%) being the most common drug related AEs at the MTD in Part 1 and 2, respectively. Most AEs were low grade. Across all cohorts, cytokine release syndrome (CRS) (G2 n=4, G3 n=2) occurred in 6 (Part 1 n=2; Part 2 n=4) and pneumonitis (G3) occurred in 5 (Part 1 n=4; Part 2 n=1) patients. Immune activation in the TME consistent with CD40 agonism and increases serum inflammatory cytokines were observed. Evidence of anti-tumor activity/clinical benefit include SD (n=13), tumor cavitation (n=2) and a uPR in solid tumors. A patient with follicular lymphoma has an ongoing durable complete metabolic response. Conclusions The CDX-1140 MTD dose of 1.5 mg/kg, a dose level expected to provide good systemic exposure and TME penetration, is generally well tolerated alone and with CDX-301. Transaminitis and CRS have generally been low grade and infrequent. A cohort combining CDX-1140 with chemotherapy will be initiated in patients with previously untreated metastatic pancreatic adenocarcinoma. Trial Registration NCT03329950 Ethics Approval The study was approved by the following: Providence St. Joseph Health IRB, approval number MOD2020001128; WIRB, approval number 1188814 (Hauke, Gabrail, Bordoni & Gordon); University of Pennsylvania IRB, approval number UPCC 18917; Mount Sinai School of Medicine IRB, approval number 18-00202; Memorial Sloan Kettering Cancer Center IRB, approval number 18-225A; Houston Methodist IRB, approval number MOD00000836
- Published
- 2020
22. 281 JAVELIN Medley VEGF: phase 2 study of avelumab + axitinib in patients with previously treated non-small cell lung cancer (NSCLC) or treatment naive, cisplatin-ineligible urothelial cancer (UC)
- Author
-
Stephen Dyar, Zsolt Horvath, David Lorente Estelles, Elena Poddubskaya, Ji-Youn Han, Viran Holden, Allison Goldman, Gabriella Galffy, Wu Chou Su, Abhimanyu Ghose, Jing Wang, Dae Ho Lee, Mikhail Laskov, Alessandra di Pietro, Ralph J. Hauke, Myung-Ju Ahn, Yu Jung Kim, Iwona Lugowska, Byoung Chul Cho, Piotr Serwatowski, Vladimir Vladimirov, and Danielle Murphy
- Subjects
Oncology ,medicine.medical_specialty ,Metastatic Urothelial Carcinoma ,business.industry ,Perforation (oil well) ,Phases of clinical research ,non-small cell lung cancer (NSCLC) ,medicine.disease ,Avelumab ,Axitinib ,Internal medicine ,Cohort ,medicine ,Lung cancer ,business ,medicine.drug - Abstract
Background Avelumab, a human anti–PD-L1 monoclonal antibody, has shown a manageable safety profile and antitumor activity in multiple tumor types, including platinum-resistant metastatic or recurrent NSCLC,1 and is approved for patients with locally advanced or metastatic UC who have progressed after ≥1 previous line of platinum-based chemotherapy2 3 and as maintenance treatment for those who have not progressed with platinum-based chemotherapy.4 JAVELIN Medley VEGF (NCT03472560) evaluated the efficacy and safety of avelumab + axitinib, a potent inhibitor of VEGFR 1, 2, and 3, in patients with advanced or metastatic NSCLC or UC. Methods Eligible patients with NSCLC had received ≥1 prior platinum-containing therapy and ≤2 prior lines of systemic therapy for locally advanced or metastatic disease; patients with UC were treatment naive in the locally advanced or metastatic setting and ineligible for cisplatin-containing chemotherapy. Patients were immune checkpoint inhibitor naive and received avelumab 800 mg intravenously every 2 weeks + axitinib 5 mg orally twice daily. The primary endpoint was confirmed objective response (OR) per investigator assessment (RECIST 1.1). Secondary endpoints included progression-free survival (PFS) and safety. PD-L1 expression was assessed in baseline tumor samples (Ventana SP263 assay). Data have not undergone standard quality checks and are subject to change due to COVID-19–related healthcare burden. Results A total of 41 patients with NSCLC and 20 with UC received avelumab + axitinib. The confirmed OR rate was 31.7% (95% CI, 18.1–48.1) in the NSCLC cohort and 10% (95% CI, 1.2–31.7) in the UC cohort (all partial responses); 16 patients (39.0%) and 5 (25.0%) had stable disease, respectively. Responses were observed regardless of PD-L1 expression status. Median PFS was 5.5 months (95% CI, 2.5–7.0) in the NSCLC cohort and 2.3 months (95% CI, 1.8–5.6) in the UC cohort. Grade ≥3 treatment-related adverse events (TRAEs) occurred in 24 patients (58.5%) in the NSCLC cohort; the most common was hypertension (n=7 [17.1%]). Grade ≥3 TRAEs occurred in 9 patients (45.0%) in the UC cohort; the most common were amylase increased, asthenia, decreased appetite, and palmar-plantar erythrodysesthesia syndrome (n=2 [10%] each). One patient in each cohort experienced a TRAE that led to death (gastric perforation and urinary bladder hemorrhage). Conclusions Avelumab + axitinib showed antitumor activity and a manageable safety profile in patients with advanced or metastatic NSCLC or UC consistent with findings from studies of each drug alone and in combination. Trial Registration NCT03472560 Ethics Approval The study was approved by each site’s independent ethics committee. Consent N/A References Gulley JL, Rajan A, Spigel DR, et al. Avelumab for patients with previously treated metastatic or recurrent non-small-cell lung cancer (JAVELIN Solid Tumor): dose-expansion cohort of a multicentre, open-label, phase 1b trial. Lancet Oncol 2017;18:599–610. Patel MR, Ellerton J, Infante JR, et al. Avelumab in metastatic urothelial carcinoma after platinum failure (JAVELIN Solid Tumor): pooled results from two expansion cohorts of an open-label, phase 1 trial. Lancet Oncol 2018;19:51–64. Bavencio(avelumab) injection. [package insert] Darmstadt, Germany: Merck KGaA; 2019. US Food and Drug Administration. FDA approves avelumab for urothelial carcinoma maintenance treatment. https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-avelumab-urothelial-carcinoma-maintenance-treatment. Accessed August 19, 2020.
- Published
- 2020
23. Safety and Efficacy of Nivolumab in Patients With Advanced Non-Clear Cell Renal Cell Carcinoma: Results From the Phase IIIb/IV CheckMate 374 Study
- Author
-
Nicholas J. Vogelzang, Mark R. Olsen, Joshua J. McFarlane, Edward Arrowsmith, Todd M. Bauer, Rohit K. Jain, Bradley Somer, Elaine T. Lam, Mark D. Kochenderfer, Ana Molina, Gurjyot Doshi, Brian Lingerfelt, Ralph J. Hauke, Vijay Gunuganti, Ian Schnadig, Peter Van Veldhuizen, Mark Fleming, Robert Galamaga, Mukul Gupta, Hugo Hool, Thomas Hutson, Joshua Zhang, M. Brent McHenry, Jennifer L. Johansen, and Scott S. Tykodi
- Subjects
medicine.medical_specialty ,Urology ,030232 urology & nephrology ,Gastroenterology ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Renal cell carcinoma ,Internal medicine ,Clinical endpoint ,Medicine ,Humans ,Adverse effect ,Carcinoma, Renal Cell ,business.industry ,medicine.disease ,Rash ,Kidney Neoplasms ,Progression-Free Survival ,Clinical trial ,Regimen ,Clear cell renal cell carcinoma ,Nivolumab ,Oncology ,030220 oncology & carcinogenesis ,medicine.symptom ,business - Abstract
Background The open-label, phase IIIb/IV CheckMate 374 study (NCT02596035) was conducted to validate the safety and efficacy of flat-dose nivolumab monotherapy 240 mg every 2 weeks (Q2W) in previously treated advanced/metastatic renal cell carcinoma (RCC). Three cohorts included patients with predominantly clear cell histology, non-clear cell histologies, or brain metastases. We report safety and efficacy from the CheckMate 374 advanced clear cell RCC (ccRCC) cohort. Patients and Methods Eligible patients received prior treatment regimens (1-2 antiangiogenic; 0-3 systemic) with progression on/after last treatment and ≤ 6 months of enrollment. Patients received nivolumab 240 mg Q2W for ≤ 24 months or until confirmed progression/unacceptable toxicity. The primary endpoint was incidence of high-grade (grade 3-5) immune-mediated adverse events (IMAEs). Exploratory endpoints included objective response rate, progression-free survival, and overall survival. Results Ninety-seven patients had advanced predominantly ccRCC; 75.3% received only 1 prior systemic regimen in the advanced/metastatic setting. After a median follow-up of 17 months (range, 0.4-26.9 months), no grade 5 IMAEs occurred, and 9.3% of patients reported grade 3/4 IMAEs (hepatitis, 4.1%; diabetes mellitus, 2.1%; nephritis and renal dysfunction, 1.0%; rash, 1.0%; adrenal insufficiency, 1.0%). The objective response rate was 22.7% (95% confidence interval [CI], 14.8%-32.3%). Three patients had a complete response; 19 had partial responses. The median progression-free survival was 3.6 months (95% CI, 2.0-5.5 months). The median overall survival was 21.8 months (95% CI, 17.4 months to not estimable). Conclusions This study validates the safety and efficacy of nivolumab 240 mg Q2W flat-dose monotherapy for previously treated advanced ccRCC and adds to previous safety and efficacy data using the 3 mg/kg Q2W dose.
- Published
- 2020
24. Safety and Efficacy of Nivolumab in Patients With Advanced Clear Cell Renal Cell Carcinoma: Results From the Phase IIIb/IV CheckMate 374 Study
- Author
-
Joshua J. McFarlane, Mark D. Kochenderfer, Mark R. Olsen, Todd M. Bauer, Ana Molina, Ralph J. Hauke, James A. Reeves, Sunil Babu, Peter Van Veldhuizen, Bradley Somer, Vijay Gunuganti, Ian Schnadig, Saby George, Ray D. Page, Edward Arrowsmith, Rohit K. Jain, Joshua Zhang, M. Brent McHenry, Jennifer L. Johansen, and Nicholas J. Vogelzang
- Subjects
Cohort Studies ,Nivolumab ,Oncology ,Urology ,Humans ,Carcinoma, Renal Cell ,Kidney Neoplasms ,Progression-Free Survival - Abstract
The open-label, phase IIIb/IV CheckMate 374 study (NCT02596035) was conducted to validate the safety and efficacy of flat-dose nivolumab monotherapy 240 mg every 2 weeks (Q2W) in previously treated advanced/metastatic renal cell carcinoma (RCC). Three cohorts included patients with predominantly clear cell histology, non-clear cell histologies, or brain metastases. We report safety and efficacy from the CheckMate 374 advanced clear cell RCC (ccRCC) cohort.Eligible patients received prior treatment regimens (1-2 antiangiogenic; 0-3 systemic) with progression on/after last treatment and ≤ 6 months of enrollment. Patients received nivolumab 240 mg Q2W for ≤ 24 months or until confirmed progression/unacceptable toxicity. The primary endpoint was incidence of high-grade (grade 3-5) immune-mediated adverse events (IMAEs). Exploratory endpoints included objective response rate, progression-free survival, and overall survival.Ninety-seven patients had advanced predominantly ccRCC; 75.3% received only 1 prior systemic regimen in the advanced/metastatic setting. After a median follow-up of 17 months (range, 0.4-26.9 months), no grade 5 IMAEs occurred, and 9.3% of patients reported grade 3/4 IMAEs (hepatitis, 4.1%; diabetes mellitus, 2.1%; nephritis and renal dysfunction, 1.0%; rash, 1.0%; adrenal insufficiency, 1.0%). The objective response rate was 22.7% (95% confidence interval [CI], 14.8%-32.3%). Three patients had a complete response; 19 had partial responses. The median progression-free survival was 3.6 months (95% CI, 2.0-5.5 months). The median overall survival was 21.8 months (95% CI, 17.4 months to not estimable).This study validates the safety and efficacy of nivolumab 240 mg Q2W flat-dose monotherapy for previously treated advanced ccRCC and adds to previous safety and efficacy data using the 3 mg/kg Q2W dose.
- Published
- 2020
25. Safety and Antitumor Activity of Apalutamide (ARN-509) in Metastatic Castration-Resistant Prostate Cancer with and without Prior Abiraterone Acetate and Prednisone
- Author
-
Rajesh Bandekar, Ralph J. Hauke, Joshi J. Alumkal, Neal D. Shore, Dana E. Rathkopf, Margaret K. Yu, Emmanuel S. Antonarakis, Mansoor N. Saleh, Charles J. Ryan, Carla de Boer, Ronald F. Tutrone, Howard I. Scher, and Edna Chow Maneval
- Subjects
Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Abiraterone Acetate ,Urology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,0302 clinical medicine ,Prednisone ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Humans ,Neoplasm Metastasis ,Adverse effect ,Aged ,Aged, 80 and over ,Chemotherapy ,business.industry ,Apalutamide ,Abiraterone acetate ,Cancer ,Middle Aged ,Prostate-Specific Antigen ,medicine.disease ,Surgery ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,Thiohydantoins ,Oncology ,chemistry ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,business ,medicine.drug - Abstract
Purpose: To evaluate the efficacy of apalutamide before or after treatment with abiraterone acetate and prednisone (AAP) in patients with progressive metastatic castration-resistant prostate cancer (mCRPC). Experimental Design: Two cohorts were studied: AAP-naïve and post-AAP patients who had received ≥6 months of AAP. Patients had progressive mCRPC per rising prostate-specific antigen (PSA) and/or imaging, without prior chemotherapy exposure. All received apalutamide 240 mg/day. Primary endpoint was ≥50% decline in 12-week PSA according to Prostate Cancer Working Group 2 criteria. Secondary endpoints included time to PSA progression and time on treatment. Results: Forty-six patients enrolled in the AAP-naïve (n = 25) and post-AAP (n = 21) cohorts. The 12-week PSA response rate was 88% (22/25) and 22% (4/18), median time to PSA progression was 18.2 months [95% confidence interval (CI), 8.3 months–not reached) and 3.7 months (95% CI, 2.8–5.6 months), and median time on treatment 21 months (range, 2.6–37.5) and 4.9 months (range, 1.3–23.2), for the AAP-naïve and post-AAP cohorts, respectively. Eighty percent (95% CI, 59–93) and 64% (95% CI, 43–82) of AAP-naïve and 43% (95% CI, 22–66) and 10% (95% CI, 1–30) of post-AAP patients remained on treatment for 6+ and 12+ months, respectively. Common treatment-emergent adverse events in both cohorts were grade 1 or 2 fatigue, diarrhea, nausea, and abdominal pain. Conclusions: Apalutamide was safe, well tolerated, and demonstrated clinical activity in mCRPC, with 80% of AAP-naïve and 43% of post-AAP patients, remaining on treatment for 6 months or longer. Clin Cancer Res; 23(14); 3544–51. ©2017 AACR.
- Published
- 2017
26. A phase 2, randomized trial evaluating the combination of dalantercept plus axitinib in advanced clear cell renal cell carcinoma
- Author
-
Robert S. Alter, J. Thaddeus Beck, Monika Joshi, Earle F. Burgess, David R. Shaffer, Mayer Fishman, Ralph J. Hauke, Nicholas J. Vogelzang, Martin H. Voss, Nauman Moazzam, Xiaosha Zhang, Chad E. Glasser, Elizabeth R. Plimack, Rupal S. Bhatt, Michael B. Atkins, Theodore F. Logan, Rahul A. Parikh, Matthew L. Sherman, and Brian I. Rini
- Subjects
Adult ,Diarrhea ,Male ,Cancer Research ,medicine.medical_specialty ,Randomization ,Axitinib ,Activin Receptors, Type II ,Recombinant Fusion Proteins ,Urology ,Angiogenesis Inhibitors ,Placebo ,Article ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,Double-Blind Method ,Renal cell carcinoma ,law ,Antineoplastic Combined Chemotherapy Protocols ,Clinical endpoint ,Medicine ,Humans ,030212 general & internal medicine ,Carcinoma, Renal Cell ,Fatigue ,Aged ,Aged, 80 and over ,business.industry ,Hazard ratio ,Middle Aged ,medicine.disease ,Kidney Neoplasms ,Progression-Free Survival ,Immunoglobulin Fc Fragments ,Clear cell renal cell carcinoma ,Oncology ,030220 oncology & carcinogenesis ,Hypertension ,Female ,business ,medicine.drug - Abstract
BACKGROUND In a prior open-label study, the combination of dalantercept, a novel antiangiogenic targeting activin receptor-like kinase 1 (ALK1), plus axitinib was deemed safe and tolerable with a promising efficacy signal in patients with advanced renal cell carcinoma (RCC). METHODS In the current phase 2, randomized, double-blind, placebo-controlled study, patients with clear cell RCC previously treated with 1 prior angiogenesis inhibitor were randomized 1:1 to receive axitinib plus dalantercept versus axitinib plus placebo. Randomization was stratified by the type of prior therapy. The primary endpoint was progression-free survival (PFS). Secondary endpoints were PFS in patients with ≥2 prior lines of anticancer therapy, overall survival, and the objective response rate. RESULTS Between June 10, 2014, and February 23, 2017, a total of 124 patients were randomly assigned to receive axitinib plus dalantercept (59 patients) or placebo (65 patients). The median PFS was not found to be significantly different between the treatment groups (median, 6.8 months vs 5.6 months; hazard ratio, 1.11 [95% CI, 0.71-1.73; P = .670]). Neither group reached the median overall survival (hazard ratio, 1.39 [95% CI, 0.70-2.77; P = .349]). The objective response rate was 19.0% (11 of 58 patients; 95% CI, 9.9%-31.4%) in the dalantercept plus axitinib group and 24.6% (15 of 61 patients; 95% CI, 14.5%-37.3%) in the placebo plus axitinib group. At least 1 treatment-emergent adverse event of ≥grade 3 was observed in 59% of patients (34 of 58 patients) in the dalantercept group and 64% of patients (39 of 61 patients) in the placebo group. One treatment-related death occurred in the placebo plus axitinib group. CONCLUSIONS Although well tolerated, the addition of dalantercept to axitinib did not appear to improve treatment-related outcomes in previously treated patients with advanced RCC.
- Published
- 2019
27. Overall survival by clinical risk category for high dose interleukin-2 (HD IL-2) treated patients with metastatic renal cell cancer (mRCC): data from the PROCLAIMSM registry
- Author
-
Scott S. Tykodi, Brendan Curti, J. Treisman, Neeraj Agarwal, Nancy C. Gregory, Janice P. Dutcher, Gregory A. Daniels, Michael Ka Keu Wong, Gerald P Miletello, David F. McDermott, Ralph J. Hauke, Michael A. Morse, Howard L. Kaufman, Helen Moon, Joseph I. Clark, Ajjai Alva, Kathleen M. Mahoney, and Mayer Fishman
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,Survival ,medicine.medical_treatment ,Tyrosine-kinase inhibitor ,0302 clinical medicine ,Renal cell carcinoma ,Aldesleukin ,Risk of mortality ,Immunology and Allergy ,Molecular Targeted Therapy ,Prospective Studies ,Neoplasm Metastasis ,Renal cell cancer ,Middle Aged ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Kidney Neoplasms ,Prior Therapy ,Treatment Outcome ,030220 oncology & carcinogenesis ,Molecular Medicine ,Patient registry ,Female ,medicine.drug ,Research Article ,Interleukin 2 ,medicine.medical_specialty ,medicine.drug_class ,Immunology ,Antineoplastic Agents ,lcsh:RC254-282 ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,Risk factor ,Carcinoma, Renal Cell ,Aged ,Retrospective Studies ,Pharmacology ,Dose-Response Relationship, Drug ,business.industry ,Immunotherapy ,medicine.disease ,PROCLAIMSM ,Survival Analysis ,030104 developmental biology ,Risk factors ,Interleukin-2 ,business - Abstract
Background Prognostic scoring systems are used to estimate the risk of mortality from metastatic renal cell carcinoma (mRCC). Outcomes from different therapies may vary within each risk group. These survival algorithms have been applied to assess outcomes in patients receiving T-cell checkpoint inhibitory immunotherapy and tyrosine kinase inhibitor therapy, but have not been applied extensively to patients receiving high dose interleukin-2 (HD IL-2) immunotherapy. Methods Survival of 810 mRCC patients treated from 2006 to 2017 with high dose IL-2 (aldesleukin) and enrolled in the PROCLAIMSM registry data base was assessed utilizing the International Metastatic RCC Database Consortium (IMDC) risk criteria. Median follow-up is 23.4 months (mo.) (range 0.2–124 mo.). Subgroup evaluations were performed by separating patients by prior or no prior therapy, IL-2 alone, or therapy subsequent to IL-2. Some patients were in two groups. We will focus on the 356 patients who received IL-2 alone, and evaluate outcome by risk factor categories. Results Among the 810 patients, 721 were treatment-naïve (89%) and 59% were intermediate risk. Overall, of the 249 patients with favorable risk, the median overall survival (OS) is 63.3 mo. and the 2-year OS is 77.6%. Of 480 patients with intermediate risk, median OS is 42.4 mo., 2-year OS 68.2%, and of 81 patients with poor risk, median OS 14 mo., 2-year OS 40.4%. Among those who received IL-2 alone (356 patients), median OS is 64.5, 57.6, and 14 months for favorable, intermediate and poor risk categories respectively. Two year survival among those treated only with HD IL-2 is 73.4, 63.7 and 39.8%, for favorable, intermediate and poor risk categories respectively. Conclusions Among mRCC patients treated with HD IL-2, all risk groups have median and 2-year survival consistent with recent reports of checkpoint or targeted therapies for mRCC. Favorable and intermediate risk (by IMDC) patients treated with HD IL-2 have longer OS compared with poor risk patients, with most durable OS observed in favorable risk patients. Favorable risk patients treated with HD IL-2 alone have a 2-year OS of 74%. These data continue to support a recommendation for HD IL-2 for patients with mRCC who meet eligibility criteria. Trial registration PROCLAIM, NCT01415167 was registered with ClinicalTrials.gov on August 11, 2011, and initiated for retrospective data collection until 2006, and prospective data collection ongoing since 2011. Electronic supplementary material The online version of this article (10.1186/s40425-019-0567-3) contains supplementary material, which is available to authorized users.
- Published
- 2019
28. An explorary analysis of a COVID-truncated REPLATINUM phase 3 trial in SCLC
- Author
-
Ralph J. Hauke, Scott Caroen, Jeannie Williams, Xiaohui Wang, Mary J. Fidler, Alberto Chiappori, Erica Burbano, Xiaoning Guo, Nacer Abrouk, Muhammad A. Salamat, Mary Flanagan Quinn, and Tony R. Reid
- Subjects
Cisplatin ,Oncology ,Cancer Research ,medicine.medical_specialty ,Coronavirus disease 2019 (COVID-19) ,business.industry ,Hazard ratio ,Carboplatin ,Clinical trial ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Toxicity ,Medicine ,Progression-free survival ,business ,Etoposide ,medicine.drug - Abstract
e20594 Background: RRx-001 is a small molecule immunotherapeutic in Phase 3 for the treatment of SCLC that is associated with M2-M1 tumor associated macrophage (TAM) repolarization with CD47 downregulation, vascular normalization and reversal of chemoresistance. In a controlled, multicenter clinical trial called REPLATINUM that was prematurely halted due to COVID-19, data from 17 patients with third line or beyond SCLC was analyzed. Moving forward, REPLATINUM(restarted) has added SARS-CoV-2 exclusion criteria. Our aim was to explore the efficacy of RRx-001 in REPLATINUM and to assess the statistical assumptions of the REPLATINUM(restarted) trial in order to inform future clinical development in SCLC. Methods: Patients in REPLATINUM were randomized to receive 1 of 2 arms: 1) carboplatin AUC 5 IV on day 1 or cisplatin 60 mg/m2 IV on day 1 plus etoposide 100 mg/m2 IV on days 1 through 3 every 21 days for up to 4 cycles or 2) 4 mg of RRx-001 administered sequentially with 4 cycles of a platinum doublet (cisplatin or carboplatin plus etoposide as outlined above). Progression Free Survival (PFS) was the primary efficacy endpoint based on a blinded independent central review (BICR). Results: The trial was suspended prematurely after 17 patients had been enrolled due to widespread COVID-19 exposure with the plan to restart the trial. The decision to restart the trial has provided an opportunity to examine the data for preliminary evidence of treatment efficacy. At the time that the REPLATINUM trial was halted, there were 11 patients enrolled on the control arm and 6 on the investigational arm. The BICR assessed median PFS was approximately 7.1 months for RRx-001 + platinum doublet and 3.5 months for the platinum doublet alone based on the truncated database. The BICR assessed PFS hazard ratio was approximately 0.5. OS medians were approximately 8.2 and 6.3 months for RRx-001 + platinum doublet and platinum doublet alone, respectively. Additionally, patients on the RRx-001 + platinum doublet-treated experimental arm experienced less toxicity than patients on the platinum doublet-treated control arm. Conclusions: Despite the small sample size, preliminary results from REPLATINUM suggest a trend toward a favorable primary outcome and improved safety in RRx-001-treated patients and support the validity of the statistical assumptions that underlie the REPLATINUM(restarted) trial. Pivotal evidence will emerge from REPLATINUM(restarted), which is imminently recommencing. Clinical trial information: NCT03699956.
- Published
- 2021
29. Phase II clinical study of concurrent durvalumab and radiation therapy (DUART) followed by adjuvant durvalumab in patients with localized urothelial cancer of bladder: Results for primary analyses and survival. BTCRC-GU15-023
- Author
-
David J. DeGraff, Deepak Kilari, Matthew Kaag, Yousef Zakharia, Hamid Emamekhoo, Sheldon L. Holder, Mark N. Stein, Monika Joshi, Ralph J. Hauke, Alexander Sankin, Joseph J. Drabick, Leonard Tuanquin, Joshua I. Warrick, Jason Liao, Hong Zheng, Benjamin A. Gartrell, and Suzanne B. Merrill
- Subjects
Oncology ,Cisplatin ,Cancer Research ,medicine.medical_specialty ,Durvalumab ,Bladder cancer ,business.industry ,medicine.medical_treatment ,medicine.disease ,Clinical study ,Radiation therapy ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Urothelial cancer ,In patient ,business ,Adjuvant ,030215 immunology ,medicine.drug - Abstract
398 Background: Bladder cancer (BC) patients (pts) who are cisplatin ineligible/unfit for surgery, or locally advanced and unresectable have limited treatment options. DUART investigates if the combination of radiation therapy (RT) and checkpoint inhibitor, durvalumab (durva) is safe and effective in these pts. We recently reported that the combination was safe, tolerable and disease control rate (DCR) was 92% post durvaRT. Here we present interim efficacy data of our phase II study. Methods: Pts with pure or mixed urothelial bladder cancer (T2-4 N0-2 M0) were enrolled if their tumor was unresectable (35%), were unfit for surgery (50%) and/or cisplatin ineligible (89%). Primary endpoints: a) PFS at 1-yr b) DCR post adjuvant durva; Secondary endpoints: a) CR post durvaRT b) median PFS c) median OS. Pts were treated with durva (1500mg) Q4 wks x2 doses along with definitive RT (64.8Gy, 36 fractions over 7 wks) to the bladder and involved nodes followed by adjuvant durva Q4 wks x 1 yr. Response was evaluated with CT scan and cystoscopy+biopsy. Sample size was based on assumption that this regimen would increase 1 yr PFS by 25% compared to RT alone (50% to 75%); we assumed DCR of 75%. A total of 26 pts were needed to reach a statistical power of at least 80% at one-sided alpha of 5% and to allow for 10% drop out rate. Results: Twenty-six pts (19 males, 7 females) were enrolled, median age 74 yr (51-94). Sixty two percent of pts had >T2 disease, 31% had positive lymph nodes; 62% with unresectable tumor or were unfit for surgery due to comorbidities. At data cut off (9/30/2020) 20/26 pts were evaluable for DCR post adjuvant durva (3 pts with CR post durvaRT, did not get adjuvant therapy; 1 pt withdrew after 3 cycles for adjuvant durva and was on f/u with unconfirmed CR; 2 pts are still on adjuvant durva) and 25/26 for PFS and all 26 pts for OS. Post completion of adjuvant durva, DCR was seen in 70 % (14/20 with 10 CR; 3 PR; 1 SD; 6 PD). One-year probability of PFS was 73% (95% CI 56.4%, 94.4%), median PFS was 18.5 months. One-year OS probability was 83.8% (95% CI 70.4%, 99.7%) with two-year OS probability of 76.8 (95% CI 60.2%, 98%). Median OS has not been reached. We did not observe any correlation between clinical outcome and baseline tumor PD-L1 expression. Conclusions: DurvaRT followed by adjuvant durva demonstrated promising efficacy with 1-year PFS probability of 73%, 1- year OS probability of 83.8% and DCR of 70% in MIBC and locally advanced BC pts with comorbidities. Results will be updated prior to the final presentation. Efficacy was also seen in node (+) pts which led to the design of prospective randomized NCTN study. Induction chemo followed by chemo+durvaRT+ adjuvant durva vs. chemoRT combination is being evaluated in the ongoing EA8185 clinical trial (ECOG-ACRIN/NRG study) for node (+) BC pts. Clinical trial information: NCT02891161.
- Published
- 2021
30. A phase I study of capivasertib in combination with abiraterone acetate in patients with metastatic castration-resistant prostate cancer
- Author
-
Rana Anjum, Satish Shah, Neal D. Shore, Thomas Morris, Zsolt Szijgyarto, Marie Cullberg, Remy B Verheijen, Ralph J. Hauke, Begoña Mellado, and Dan Costin
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Abiraterone acetate ,Castration resistant ,medicine.disease ,Phase i study ,Androgen receptor ,chemistry.chemical_compound ,Prostate cancer ,chemistry ,Internal medicine ,Medicine ,In patient ,business - Abstract
85 Background: Androgen receptor (AR) targeting therapies prolong survival of patients with metastatic castration-resistant prostate cancer (mCRPC); however, in many cases, resistance develops, resulting in disease progression. Activation of the PI3K/AKT/mTOR signaling pathway is common in mCRPC and contributes to resistance, mostly due to loss of PTEN, which occurs in 40–60% of patients. Preclinical studies have demonstrated reciprocal regulation between the AR and PI3K/AKT/mTOR pathways and significant anti-tumor activity when both pathways are inhibited, particularly in models with PTEN-loss. Thus, a rationale exists to inhibit both pathways in mCRPC patients. We report interim results of a phase 1 multicohort study (NCT04087174) to confirm the acceptable dose of capivasertib, a potent, selective pan-AKT inhibitor in combination with the androgen synthesis inhibitor abiraterone acetate (AA) in mCRPC patients. Methods: Patients who had received at least one prior systemic therapy (chemotherapy or novel hormonal agent) for mCRPC were given AA (1000 mg, once daily) with capivasertib (400 mg, twice daily, 4 days on/3 days off) until unacceptable toxicity or disease progression. Dose-limiting toxicity in the first 28 days of treatment and adverse events were recorded. Results: 15 patients, median age 67 (range 49–82) years, were recruited in the USA and Spain. Twelve patients had received prior chemotherapy; 7 had two or more prior lines. Seven patients had received prior AA and 10 had received prior enzalutamide. No dose-limiting toxicities were recorded. Eight patients reported at least one grade ≥ 3 adverse event (AE). Grade ≥ 3 AEs in 7 patients were related to capivasertib: allergic reaction to medication, asthenia, type 2 diabetes mellitus, diarrhea and fatigue were each reported in 1 patient, maculopapular rash − in 2 patients, both hypokalemia and acquired Fanconi syndrome − in 1 patient. Acute kidney injury was reported in 4 patients but was not considered related to capivasertib. The most common AEs of any grade related to capivasertib were: diarrhea, 6/15 patients (40%); maculopapular rash, 5/15 (33%); fatigue, 4/15 (27%); hyperglycemia/type 2 diabetes mellitus, 4/15 (27%); nausea, 3/15 (20%); hypokalemia, 2/15 (13%); hypophosphatemia, 2/15 (13%). Capivasertib was discontinued in 4/15 patients (27%) due to AEs. Between initial screening and day 29 of treatment, 5 patients had reduced (> 20%) PSA levels, with 3 patients showing sustained falls in PSA over 12 weeks. Conclusions: In this phase 1 study combined capivasertib and AA exhibits an acceptable safety and tolerability profile. Further data on the clinical efficacy and safety of the combination are being collected in the phase 3 CAPItello-281 trial. Acknowledgments: We thank Adam Errington, PhD, of Oxford PharmaGenesis, for medical writing assistance. Funding: This trial is funded by AstraZeneca. Clinical trial information: NCT04087174.
- Published
- 2021
31. Angiogenic and T-effector subgroups identified by gene expression profiling (GEP) and propensity for PBRM1 and BAP1 alterations in clear cell renal cell carcinoma (ccRCC)
- Author
-
Shuchi Gulati, Tian Zhang, Charles J. Ryan, David I. Quinn, Hans J. Hammers, Andrew Elliott, Nancy A. Dawson, Chadi Nabhan, Daniel M. Geynisman, Shuanzeng Wei, Sourat Darabi, Ralph J. Hauke, Matthew Zibelman, Pedro C. Barata, Arpit Rao, Kelsey Poorman, Elisabeth I. Heath, and Benjamin A. Gartrell
- Subjects
Cancer Research ,BAP1 ,business.industry ,Effector ,medicine.disease ,PBRM1 ,Gene expression profiling ,Clear cell renal cell carcinoma ,Oncology ,Gene expression ,medicine ,Cancer research ,Active treatment ,business ,Selection (genetic algorithm) - Abstract
343 Background: With the emergence of multiple active treatment options in RCC, predictive biomarkers for optimal treatment selection are lacking. Gene expression data from IMmotion151 and Javelin Renal 101 clinical trials generated anti-angiogenic and immune signatures that warrant further validation. We aimed to describe the genomic and gene expression profiles in a multi-institutional database of patients with ccRCC, and its association with other biomarkers of interest. Methods: Whole transcriptome sequencing was performed for ccRCC patient samples submitted to a commercial CLIA-certified laboratory (Caris Life Sciences, Phoenix, AZ) from February 2019 to September 2020. Tumor GEP and hierarchical clustering based on the validated 66-gene signature (D’Costa et al, 2020) were used to identify patient subgroups. Samples from both primary tumors and metastatic sites were included. Results: A total of 316 patients with ccRCC, median age 62 (range 32-90), 71.8% men, were included. Tissue samples were obtained from primary tumor (46.5%), lung (12.3%), bone (9.5%), liver (4.7%) and other metastatic sites (27%). Gene expression analysis identified angiogenic, mixed and T-effector subgroups in 24.1%, 51.3% and 24.7%, respectively. Patients with angiogenic subgroup tumors compared to those with T-effector subgroup tumors were more likely to be older (63 versus 60 years, p=0.035), female (40.8% versus 16.7%, p=0.0009) and more frequently found in pancreatic/small bowel metastases (75% versus 12.5%, p=0.0103). Biomarkers of potential response to immunotherapy such as PD-L1 (p=0.0021), TMB (not significant), and dMMR/MSI-H status (not significant) were more frequent in the T-effector subgroup. PBRM1 mutations were more common in the angiogenic subgroup (62.0% vs 37.5%, p=0.0034) while BAP1 mutations were more common in the T-effector subgroup (18.6% versus 3.0%, p= 0.0035). Immune cell population abundance (e.g. NK cells, monocytes) and immune checkpoint gene expression (TIM-3, PD-L1, PD-L2, CTLA4) were also increased in the T-effector subgroup. Conclusions: Our hierarchical clustering results based on the 66-gene expression signature were concordant with results from prior studies. Patient subgroups identified by evaluation of angiogenic and T-effector signature scores exhibit significantly different mutations and immune profiles. These findings require prospective validation in future biomarker-selected clinical trials.
- Published
- 2021
32. Guideline on Muscle-Invasive and Metastatic Bladder Cancer (European Association of Urology Guideline): American Society of Clinical Oncology Clinical Practice Guideline Endorsement
- Author
-
Matthew I. Milowsky, Pat Boumansour, Timothy D. Gilligan, Ralph J. Hauke, R. Bryan Rumble, Cheryl T. Lee, Libni J. Eapen, and Christopher M. Booth
- Subjects
Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,030232 urology & nephrology ,Urology ,MEDLINE ,Cystectomy ,03 medical and health sciences ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Neoplasm Metastasis ,Clinical Oncology ,business.industry ,Chemoradiotherapy ,Guideline ,Clinical Practice ,Metastatic bladder cancer ,Clinical trial ,Urinary Bladder Neoplasms ,Oncology ,030220 oncology & carcinogenesis ,Practice Guidelines as Topic ,Cisplatin ,business - Abstract
Purpose To endorse the European Association of Urology guideline on muscle-invasive (MIBC) and metastatic bladder cancer. The American Society of Clinical Oncology (ASCO) has a policy and set of procedures for endorsing clinical practice guidelines that have been developed by other professional organizations. Methods The guideline on MIBC and metastatic bladder cancer was reviewed for developmental rigor by methodologists. The ASCO Endorsement Panel then reviewed the content and recommendations. Results The ASCO Endorsement Panel determined that the recommendations from the European Association of Urology guideline on MIBC and metastatic bladder cancer, published online in March 2015, are clear, thorough, and based on the most relevant scientific evidence. ASCO endorses the guideline on MIBC and metastatic bladder cancer and has added qualifying statements, including highlighting the use of chemoradiotherapy for select patients with MIBC and recommending a preference for clinical trials in the treatment of metastatic disease in the second-line setting. Recommendations Multidisciplinary care for patients with MIBC and metastatic bladder cancer is critical. The standard treatment of MIBC (cT2-T4a N0M0) is neoadjuvant cisplatin-based combination chemotherapy followed by radical cystectomy. In cisplatin-ineligible patients, radical cystectomy alone is recommended. Adjuvant cisplatin-based chemotherapy may be offered to high-risk patients who have not received neoadjuvant therapy. Chemoradiotherapy may be offered as an alternative to cystectomy in appropriately selected patients with MIBC and in some patients for whom cystectomy is not an option. Metastatic disease should be treated with cisplatin-containing combination chemotherapy or with carboplatin combination chemotherapy or single agents in patients ineligible for cisplatin. Additional information is available at http://www.asco.org/endorsements/MIBC and www.asco.org/guidelineswiki .
- Published
- 2016
33. A phase III trial-in-progress called REPLATINUM that compares RRx-001 + a platinum doublet to a platinum doublet in third-line or beyond small cell lung cancer
- Author
-
Daniel Morgensztern, Ralph J. Hauke, John C. Ruckdeschel, Christian Rolfo, Alberto Chiappori, Tarek Mekhail, Mary J. Fidler, John T. Hamm, Konstantin H. Dragnev, Chao Hui Huang, and Raid Aljumaily
- Subjects
Cancer Research ,Poor prognosis ,business.industry ,Immune checkpoint inhibitors ,chemistry.chemical_element ,Unmet needs ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,Third line ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,Non small cell ,business ,Platinum ,Extensive-stage small cell lung cancer ,030215 immunology - Abstract
TPS9083 Background: Despite several recent checkpoint inhibitor approvals extensive stage small cell lung cancer (SCLC) is associated with a poor prognosis and remains an area of high unmet need. RRx-001 is a first-in-class, minimally toxic small molecule immunotherapeutic that inhibits c-Myc, downregulates the antiphagocytic checkpoint, CD47, repolarizes tumor associated macrophages (TAM) from protumor M2 to antitumor M1 and resensitizes to previously active first line therapies. On the basis of favorable results from a Phase 2 trial called QUADRUPLE THREAT (NCT02489903) in combination with a platinum doublet in later line SCLC, a Phase 3 trial called REPLATINUM was started in 3rd line or beyond SCLC in Q4 2019. Enrollment is ongoing. Methods: This US-based, open-label, randomized, phase 3 study (NCT03777657) compares RRx-001 4mg + a platinum doublet (carboplatin or cisplatin + etoposide) versus a platinum doublet for pts with 3rd line or beyond SCLC that have previously received a checkpoint inhibitor. Approximately 120 pts from 25 centers will be randomized 1:1 to receive RRx-001 4 mg in combination with a platinum doublet vs. a platinum doublet. The platinum doublet will be administered on both arms for up to 4 cycles; on the RRx-001 arm only patients with stable disease or better are eligible to continue on RRx-001 4 mg + carboplatin AUC 2-4 maintenance therapy. If radiologic progression occurs on the control arm prior to the 4th cycle, patients are eligible to crossover to the RRx-001 treatment arm. PFS is the primary endpoint. Secondary endpoints include OS and ORR. Exploratory endpoints include c-Myc, CD-47 and PD-L1 on circulating tumor cells and SIRP-alpha expression on circulating monocytes. Clinical trial information: NCT03777657.
- Published
- 2020
34. Phase II trial of atezolizumab plus chemotherapy after progression on single-agent PD-1 or PD-L1 inhibitor in cisplatin ineligible patients with advanced urothelial carcinoma HCRN GU17-295
- Author
-
Ralph J. Hauke, Hristos Z. Kaimakliotis, Roberto Pili, Neda Hashemi, Nabil Adra, and Shuchi Gulati
- Subjects
Cisplatin ,Cancer Research ,Chemotherapy ,Metastatic Urothelial Carcinoma ,business.industry ,medicine.medical_treatment ,Treatment options ,Oncology ,Atezolizumab ,medicine ,Cancer research ,Single agent ,business ,PD-L1 inhibitor ,medicine.drug ,Urothelial carcinoma - Abstract
TPS587 Background: Patients (pts) with cisplatin ineligible metastatic urothelial carcinoma (mUC) who progress after first-line PD1/PDL1 inhibition have limited treatment options. The concept of maintenance therapy with targeted agents and adding onto it at time of progression is a proven effective strategy. Preclinical data indicate that carboplatin+gemcitabine have immunomodulatory effect to potentially augment immune response. We hypothesize that in pts with cisplatin ineligible mUC, the use of atezolizumab+chemotherapy after progression on single-agent PD1/PDL1 inhibitor will result in clinical benefit. Methods: Multi-center, single arm, open label phase 2 trial of atezolizumab+carboplatin+gemcitabine in pts with cisplatin ineligible mUC. Eligible pts are adults with mUC (mixed histology allowed) who progressed after first line PD1/PDL1 inhibitor. Pts should be cisplatin ineligible based on consensus criteria. Neoadjuvant/adjuvant chemotherapy completed ≥12 months prior to enrollment is allowed. Treatment with atezolizumab will continue until disease progression or unacceptable toxicity while carboplatin+gemcitabine can be stopped after 4-6 cycles. Primary objective is progression-free survival per RECIST and secondary objectives are overall response rate, clinical benefit rate, and overall survival (OS). Exploratory endpoints include to compare OS of atezolizumab+carboplatin+gemcitabine in this trial compared to a virtual control arm of carboplatin+gemcitabine in mUC after progression on first-line PD1/PDL1 inhibitors. Other exploratory endpoints include to compare PD-L1 status at time of diagnosis and at time of enrollment (after progression on PD1/PDL1 inhibitor). Using an alternate hypothesis that atezolizumab+carboplatin+gemcitabine will have a median PFS of 9 months compared to historical control of 5 months with a platinum regimen in 2nd line setting, we plan to enroll 33 patients. This study is currently enrolling pts. A protocol amendment is under way that will allow pts with prior platinum-based chemotherapy to enroll on this trial. Clinical trial information: NCT03737123.
- Published
- 2020
35. Concurrent durvalumab and radiation therapy followed by adjuvant durvalumab in patients with locally advanced urothelial cancer of bladder (DUART): Btcrc-GU15-023
- Author
-
Sheldon L. Holder, Leonard Tuanquin, Joshua I. Warrick, Hong Zheng, Yousef Zakharia, Ralph J. Hauke, Alexander Sankin, Benjamin A. Gartrell, Monika Joshi, David J. DeGraff, Deepak Kilari, Matthew Kaag, Hamid Emamekhoo, Joseph J. Drabick, Mark N. Stein, and Suzanne B. Merrill
- Subjects
Cisplatin ,Oncology ,Cancer Research ,medicine.medical_specialty ,Durvalumab ,Bladder cancer ,business.industry ,medicine.medical_treatment ,Locally advanced ,medicine.disease ,Radiation therapy ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Urothelial cancer ,In patient ,business ,Adjuvant ,030215 immunology ,medicine.drug - Abstract
513 Background: Bladder cancer (BC) patients (pts) who are cisplatin ineligible/unfit for surgery, unresectable have limited treatment options. In this study, we investigate if the combination of radiation therapy (RT) and checkpoint inhibitor, durvalumab (durva) is safe and effective in these pts. Our results from phase (ph) Ib suggested that the combination was safe. Here we present the response rate post durvaRT and updated treatment related adverse events (TRAEs) amongst our evaluable pts in ph II. Methods: This is a single arm ph Ib-II study for T2-4 N0-2 M0 pts. The ph II primary endpoints a) PFS rate at 1 yr b) disease control rate (DCR); secondary endpoints were a) CR post durvaRT b) PFS c) OS. Pts were treated with durva (1500mg) Q4 wks x2 doses along with definitive RT (64.8Gy, 36 fractions over 7 wks) to the bladder and involved nodes followed by adjuvant durva Q4 wks x 1 yr. Response was evaluated with CT scan and cystoscopy+biopsy post durvaRT. We anticipated that durvaRT followed by durva would increase PFS at 1 yr from 50% to 75% when compared to RT; we assumed DCR of about 75%. A total of 26 pts were needed to reach a statistical power of at least 80% at one-sided alpha of 5% and to allow for 10% drop out rate. Results: Total N = 26 patients (male 19; female 7, median age 74yr). At the time of data cut off, 21/26 pts were evaluable for response post durvaRT. Post completion of durvaRT time point, clinical CR was seen in 15/21 pts (71.4%); PR 1/21 pts (4.7%); SD 4/21 (19%); PD 1/21 (4.7%). DCR was seen in 20/21 pts (95%) post durvaRT. Median follow up from D1 to last follow up was 6.1 mos. Grade ≥ 3 TRAE amongst 26 pts: anemia (1/26), lipase/amylase (1/26), immune nephritis (1/26), dyspnea (gr 4, copd/immune), fatigue (1/26), lymphopenia (6/26). Other TRAEs: Fatigue was the most common TRAE (16/26); UTI (5/26); cystitis (3/26). No fatal TRAEs were observed. Conclusions: DurvaRT demonstrated promising efficacy with clinical CR of 71.4% and DCR of 95% in unresectable, cisplatin ineligible locally advanced BC. It was generally well tolerated. Ph II study has completed accrual and longer-term results will further our understanding of this regimen’s efficacy in locally advanced BC. Clinical trial information: NCT02891161.
- Published
- 2020
36. Severe Hepatotoxicity of Mithramycin Therapy Caused by Altered Expression of Hepatocellular Bile Transporters
- Author
-
Julie A. Hong, Edel M. McCrea, David S. Schrump, Patrick J. Grohar, Jonathan P. Jackson, David Venzon, Mary Zhang, Kenneth R. Brouwer, William D. Figg, Ralph J. Hauke, Cody J. Peer, Jon Glod, Theo Heller, Roberto H. Barbier, Jonathan D. Strope, Tristan M. Sissung, Brigitte C. Widemann, Phoebe A. Huang, and Ariel M. Ley
- Subjects
0301 basic medicine ,Adult ,Male ,ATP Binding Cassette Transporter, Subfamily B ,medicine.drug_class ,Receptors, Cytoplasmic and Nuclear ,Bile acid binding ,Pharmacology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Clinical Trials, Phase II as Topic ,Chenodeoxycholic acid ,Cell Line, Tumor ,medicine ,Humans ,ABCB11 ,ATP Binding Cassette Transporter, Subfamily B, Member 11 ,Aged ,Antibiotics, Antineoplastic ,Bile acid ,Membrane Transport Proteins ,Plicamycin ,Articles ,Middle Aged ,Thoracic Neoplasms ,Bile Salt Export Pump ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Hepatocyte ,Small heterodimer partner ,Molecular Medicine ,Farnesoid X receptor ,Female ,Chemical and Drug Induced Liver Injury ,030217 neurology & neurosurgery - Abstract
Mithramycin demonstrates preclinical anticancer activity, but its therapeutic dose is limited by the development of hepatotoxicity that remains poorly characterized. A pharmacogenomics characterization of mithramycin-induced transaminitis revealed that hepatotoxicity is associated with germline variants in genes involved in bile disposition: ABCB4 (multidrug resistance 3) rs2302387 and ABCB11 [bile salt export pump (BSEP)] rs4668115 reduce transporter expression (P < 0.05) and were associated with ≥grade 3 transaminitis developing 24 hours after the third infusion of mithramycin (25 mcg/kg, 6 hours/infusion, every day ×7, every 28 days; P < 0.0040). A similar relationship was observed in a pediatric cohort. We therefore undertook to characterize the mechanism of mithramycin-induced acute transaminitis. As mithramycin affects cellular response to bile acid treatment by altering the expression of multiple bile transporters (e.g., ABCB4, ABCB11, sodium/taurocholate cotransporting polypeptide, organic solute transporter α/β) in several cell lines [Huh7, HepaRG, HepaRG BSEP (-/-)] and primary human hepatocytes, we hypothesized that mithramycin inhibited bile-mediated activation of the farnesoid X receptor (FXR). FXR was downregulated in all hepatocyte cell lines and primary human hepatocytes (P < 0.0001), and mithramycin inhibited chenodeoxycholic acid- and GW4046-induced FXR-galactose-induced gene 4 luciferase reporter activity (P < 0.001). Mithramycin promoted glycochenodeoxycholic acid-induced cytotoxicity in ABCB11 (-/-) cells and increased the overall intracellular concentration of bile acids in primary human hepatocytes grown in sandwich culture (P < 0.01). Mithramycin is a FXR expression and FXR transactivation inhibitor that inhibits bile flow and potentiates bile-induced cellular toxicity, particularly in cells with low ABCB11 function. These results suggest that mithramycin causes hepatotoxicity through derangement of bile acid disposition; results also suggest that pharmacogenomic markers may be useful to identify patients who may tolerate higher mithramycin doses. SIGNIFICANCE STATEMENT: The present study characterizes a novel mechanism of drug-induced hepatotoxicity in which mithramycin not only alters farnesoid X receptor (FXR) and small heterodimer partner gene expression but also inhibits bile acid binding to FXR, resulting in deregulation of cellular bile homeostasis. Two novel single-nucleotide polymorphisms in bile flow transporters are associated with mithramycin-induced liver function test elevations, and the present results are the rationale for a genotype-directed clinical trial using mithramycin in patients with thoracic malignancies.
- Published
- 2018
37. A Phase I/II Trial of BNC105P with Everolimus in Metastatic Renal Cell Carcinoma
- Author
-
Richard C. Lauer, Ravindran Kanesvaran, Gabriel Kremmidiotis, Harry A. Drabkin, Thomas E. Hutson, Jose Iglesias, Timothy Breen, Alexander Starodub, Christopher Sweeney, Stephen Lane Richey, Brian A. Costello, Shailender Bhatia, David C. Bibby, Jeremy Simpson, Annabell F. Leske, Sumanta K. Pal, Ralph J. Hauke, Elizabeth E. Doolin, Arun Azad, Guru Sonpavde, Noah M. Hahn, and John Sarantopoulos
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Phases of clinical research ,Kaplan-Meier Estimate ,Pharmacology ,Article ,Disease-Free Survival ,law.invention ,Randomized controlled trial ,law ,Renal cell carcinoma ,Sex Hormone-Binding Globulin ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Carcinoma ,Humans ,Everolimus ,Carcinoma, Renal Cell ,Aged ,Benzofurans ,Aged, 80 and over ,business.industry ,Middle Aged ,medicine.disease ,Organophosphates ,Clinical trial ,Treatment Outcome ,Matrix Metalloproteinase 9 ,Biomarker (medicine) ,Female ,business ,medicine.drug - Abstract
Purpose: BNC105P inhibits tubulin polymerization, and preclinical studies suggest possible synergy with everolimus. In this phase I/II study, efficacy and safety of the combination were explored in patients with metastatic renal cell carcinoma (mRCC). Experimental Design: A phase I study in patients with clear cell mRCC and any prior number of therapies was conducted using a classical 3 + 3 design to evaluate standard doses of everolimus with increasing doses of BNC105P. At the recommended phase II dose (RP2D), patients with clear cell mRCC and one to two prior therapies (including ≥ 1 VEGF-TKI) were randomized to BNC105P with everolimus (arm A) or everolimus alone (arm B). The primary endpoint of the study was 6-month progression-free survival (6MPFS). Secondary endpoints included response rate, PFS, overall survival, and exploratory biomarker analyses. Results: In the phase I study (N = 15), a dose of BNC105P at 16 mg/m2 with everolimus at 10 mg daily was identified as the RP2D. In the phase II study, 139 patients were randomized, with 69 and 67 evaluable patients in arms A and B, respectively. 6MPFS was similar in the treatment arms (arm A: 33.82% vs. arm B: 30.30%, P = 0.66) and no difference in median PFS was observed (arm A: 4.7 mos vs. arm B: 4.1 mos; P = 0.49). Changes in matrix metalloproteinase-9, stem cell factor, sex hormone-binding globulin, and serum amyloid A protein were associated with clinical outcome with BNC105P. Conclusions: Although the primary endpoint was not met in an unselected population, correlative studies suggest several biomarkers that warrant further prospective evaluation. Clin Cancer Res; 21(15); 3420–7. ©2015 AACR.
- Published
- 2015
38. Prostate Cancer Survivorship Care Guideline: American Society of Clinical Oncology Clinical Practice Guideline Endorsement
- Author
-
David F. Penson, Matthew J. Resnick, Karen E. Hoffman, Ralph J. Hauke, Alicia K. Morgans, Jonathan Bergman, Christina Lacchetti, and Terrence M. Kungel
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,business.industry ,Prostate ,Primary care physician ,Prostatic Neoplasms ,Cancer ,Guideline ,Medical Oncology ,medicine.disease ,United States ,Survival Rate ,Prostate cancer ,Quality of life (healthcare) ,Health promotion ,Oncology ,Survivorship curve ,Family medicine ,Practice Guidelines as Topic ,medicine ,Humans ,business ,Psychosocial ,Societies, Medical - Abstract
Purpose The guideline aims to optimize health and quality of life for the post-treatment prostate cancer survivor by comprehensively addressing components of follow-up care, including health promotion, prostate cancer surveillance, screening for new cancers, long-term and late functional effects of the disease and its treatment, psychosocial issues, and coordination of care between the survivor's primary care physician and prostate cancer specialist. Methods The American Cancer Society (ACS) Prostate Cancer Survivorship Care Guidelines were reviewed for developmental rigor by methodologists. The American Society of Clinical Oncology (ASCO) Endorsement Panel reviewed the content and recommendations, offering modifications and/or qualifying statements when deemed necessary. Results The ASCO Endorsement Panel determined that the recommendations from the 2014 ACS Prostate Cancer Survivorship Care Guidelines are clear, thorough, and relevant, despite the limited availability of high-quality evidence to support many of the recommendations. ASCO endorses the ACS Prostate Cancer Survivorship Care Guidelines, with a number of qualifying statements and modifications. Recommendations Assess information needs related to prostate cancer, prostate cancer treatment, adverse effects, and other health concerns and provide or refer survivors to appropriate resources. Measure prostate-specific antigen (PSA) level every 6 to 12 months for the first 5 years and then annually, considering more frequent evaluation in men at high risk for recurrence and in candidates for salvage therapy. Refer survivors with elevated or increasing PSA levels back to their primary treating physician for evaluation and management. Adhere to ACS guidelines for the early detection of cancer. Assess and manage physical and psychosocial effects of prostate cancer and its treatment. Annually assess for the presence of long-term or late effects of prostate cancer and its treatment.
- Published
- 2015
39. Phase 2 Trial of Gemcitabine, Cisplatin, plus Ipilimumab in Patients with Metastatic Urothelial Cancer and Impact of DNA Damage Response Gene Mutations on Outcomes
- Author
-
Bojan Losic, Guru Sonpavde, Ralph J. Hauke, Noah M. Hahn, Menggang Yu, Sacha Gnjatic, Eric E. Schadt, Vaibhav G. Patel, Nicholas K. Akers, Przemyslaw Twardowski, Josep Domingo-Domenech, Seunghee Kim-Schulze, Sumanta K. Pal, Alexander Starodub, Michael J. Donovan, Nina Bhardwaj, Costantine Albany, Mark D. Fleming, Qianqian Zhao, William Oh, Huan Wang, John P. Sfakianos, Andrew V. Uzilov, Matthew D. Galsky, and Rong Chen
- Subjects
0301 basic medicine ,Oncology ,Male ,medicine.medical_specialty ,Urologic Neoplasms ,Urology ,medicine.medical_treatment ,DNA Mutational Analysis ,Ipilimumab ,Kaplan-Meier Estimate ,Gene mutation ,Deoxycytidine ,Risk Assessment ,Disease-Free Survival ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Humans ,Neoplasm Invasiveness ,Molecular Targeted Therapy ,Prospective Studies ,Neoplasm Metastasis ,Survival analysis ,Aged ,Neoplasm Staging ,Aged, 80 and over ,Carcinoma, Transitional Cell ,business.industry ,Immunotherapy ,Middle Aged ,Prognosis ,Survival Analysis ,Gemcitabine ,Immune checkpoint ,030104 developmental biology ,Treatment Outcome ,CTLA-4 ,030220 oncology & carcinogenesis ,Mutation ,Female ,Cisplatin ,business ,medicine.drug ,DNA Damage - Abstract
Background Chemotherapy may exert immunomodulatory effects, thereby combining favorably with the immune checkpoint blockade. The pharmacodynamic effects of such combinations, and potential predictive biomarkers, remain unexplored. Objective To determine the safety, efficacy, and immunomodulatory effects of gemcitabine and cisplatin (GC) plus ipilimumab and explore the impact of somatic DNA damage response gene alterations on antitumor activity. Design, setting, and participants Multicenter single arm phase 2 study enrolling 36 chemotherapy-naive patients with metastatic urothelial cancer. Peripheral blood flow cytometry was performed serially on all patients and whole exome sequencing of archival tumor tissue was performed on 28/36 patients. Intervention Two cycles of GC followed by four cycles of GC plus ipilimumab. Outcome measurements and statistical analysis The primary endpoint was 1-yr overall survival (OS). Secondary endpoints included safety, objective response rate, and progression-free survival. Results and limitations Grade ≥3 adverse events occurred in 81% of patients, the majority of which were hematologic. The objective response rate was 69% and 1-yr OS was 61% (lower bound 90% confidence interval: 51%). On exploratory analysis, there were no significant changes in the composition and frequency of circulating immune cells after GC alone. However, there was a significant expansion of circulating CD4 cells with the addition of ipilimumab which correlated with improved survival. The response rate was significantly higher in patients with deleterious somatic DNA damage response mutations (sensitivity=47.6%, specificity=100%, positive predictive value=100%, and negative predictive value=38.9%). Limitations are related to the sample size and single-arm design. Conclusions GC+ipilimumab did not achieve the primary endpoint of a lower bound of the 90% confidence interval for 1-yr OS of >60%. However, within the context of a small single-arm trial, the results may inform current approaches combining chemotherapy plus immunotherapy from the standpoint of feasibility, appropriate cytotoxic backbones , and potential predictive biomarkers. Trial registration: ClinicalTrials.gov NCT01524991. Patient summary Combining chemotherapy and immune checkpoint blockade in patients with metastatic urothelial cancer is feasible. Further studies are needed to refine optimal combinations and evaluate tests that might identify patients most likely to benefit.
- Published
- 2017
40. Discussing the predictive, prognostic, and therapeutic value of germline DNA-repair gene mutations in metastatic prostate cancer patients
- Author
-
Ralph J. Hauke, William D. Figg, and Tristan M. Sissung
- Subjects
Pharmacology ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Journal Club ,Cancer ,Gene mutation ,Malignancy ,medicine.disease ,Precision medicine ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Germline mutation ,030220 oncology & carcinogenesis ,Internal medicine ,Epidemiology of cancer ,medicine ,Molecular Medicine ,030212 general & internal medicine ,business ,Genetic testing - Abstract
Recent trends in cancer therapy have begun emphasizing the use of precision medicine, especially genetic tools, in the evaluation of malignancies and decision-making. Prostate cancer is a malignancy where the benefits and utility of screening and early treatment are still heavily controversial. A recent paper in the New England Journal of Medicine found that patients with metastatic prostate cancer presented germline mutations in DNA-repair genes at a significantly higher incidence than those with localized prostate cancer. These findings indicate the need for further research in this field as genetic differences between metastatic and localized prostate cancer could have great clinical value.
- Published
- 2017
41. Androgen receptor mutations in patients with castration-resistant prostate cancer treated with apalutamide
- Author
-
Glenn Liu, William R. Berry, A. Trinh, Mansoor N. Saleh, Dana E. Rathkopf, Rajesh Bandekar, Charles J. Ryan, Margaret K. Yu, E. Chow Maneval, Shibu Thomas, Joshi J. Alumkal, Emmanuel S. Antonarakis, C. J. de Boer, Thian Kheoh, Howard I. Scher, Deborah Ricci, N.D. Shore, Celestia S. Higano, Ronald F. Tutrone, Ralph J. Hauke, and Matthew R. Smith
- Subjects
0301 basic medicine ,Oncology ,Male ,medicine.medical_specialty ,Urology ,medicine.disease_cause ,Circulating Tumor DNA ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,0302 clinical medicine ,Prednisone ,Internal medicine ,medicine ,Humans ,Point Mutation ,Digital polymerase chain reaction ,Aged ,Aged, 80 and over ,Mutation ,business.industry ,Apalutamide ,Disease progression ,Androgen Antagonists ,Hematology ,Original Articles ,Middle Aged ,medicine.disease ,Androgen receptor ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,chemistry ,Thiohydantoins ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Mutation testing ,business ,medicine.drug - Abstract
Background Mutations in the androgen receptor (AR) ligand-binding domain (LBD), such as F877L and T878A, have been associated with resistance to next-generation AR-directed therapies. ARN-509-001 was a phase I/II study that evaluated apalutamide activity in castration-resistant prostate cancer (CRPC). Here, we evaluated the type and frequency of 11 relevant AR-LBD mutations in apalutamide-treated CRPC patients. Patients and methods Blood samples from men with nonmetastatic CRPC (nmCRPC) and metastatic CRPC (mCRPC) pre- or post-abiraterone acetate and prednisone (AAP) treatment (≥6 months’ exposure) were evaluated at baseline and disease progression in trial ARN-509-001. Mutations were detected in circulating tumor DNA using a digital polymerase chain reaction-based method known as BEAMing (beads, emulsification, amplification and magnetics) (Sysmex Inostics’ GmbH). Results Of the 97 total patients, 51 had nmCRPC, 25 had AAP-naive mCRPC, and 21 had post-AAP mCRPC. Ninety-three were assessable for the mutation analysis at baseline and 82 of the 93 at progression. The overall frequency of detected AR mutations at baseline was 7/93 (7.5%) and at progression was 6/82 (7.3%). Three of the 82 (3.7%) mCRPC patients (2 AAP-naive and 1 post-AAP) acquired AR F877L during apalutamide treatment. At baseline, 3 of the 93 (3.2%) post-AAP patients had detectable AR T878A, which was lost after apalutamide treatment in 1 patient who continued apalutamide treatment for 12 months. Conclusions The overall frequency of detected mutations at baseline (7.5%) and progression (7.3%) using the sensitive BEAMing assay was low, suggesting that, based on this assay, AR-LBD mutations such as F877L and T878A are not common contributors to de novo or acquired resistance to apalutamide. ClinicalTrials.gov identifier NCT01171898.
- Published
- 2017
42. Immunological basis in the pathogenesis and treatment of bladder cancer
- Author
-
Michael Feloney, Larry E. Siref, Ralph J Hauke, David B Thompson, and Devendra K. Agrawal
- Subjects
alpha-Defensins ,medicine.medical_treatment ,Immunology ,Inflammation ,Cancer Vaccines ,Article ,Immune system ,Leukocytes ,medicine ,Humans ,Immunology and Allergy ,Urothelium ,Interleukin 6 ,Bladder cancer ,biology ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,business.industry ,Cancer ,medicine.disease ,Mycobacterium bovis ,Cytokine ,Urinary Bladder Neoplasms ,biology.protein ,Tumor necrosis factor alpha ,medicine.symptom ,business - Abstract
The pathogenesis and transition of normal urothelium into bladder carcinoma are multifactorial processes. Chronic inflammation causes initiation and progression of the underlying pathophysiology of invasive and metastatic cancer. A dichotomy is observed in the role of immune cells in bladder cancer. While the immune response defends the host by suppressing neoplastic growth, several immune cells, including neutrophils, macrophages, and T-lymphocytes, promote tumor development and progression. The levels of human neutrophil peptide-1, -2, and -3, produced by neutrophils, increase in bladder cancer and might promote tumor angiogenesis and growth. The effect of macrophages is primarily mediated by pro-inflammatory cytokines, IL-6 and TNF-α. Additionally, the underlying immunological mechanisms of two treatments, BCG and cytokine gene-modified tumor vaccines, and future directions are critically discussed.
- Published
- 2014
43. Immunological effects and therapeutic role of C5a in cancer
- Author
-
Devendra K. Agrawal, Victoria R Darling, Ralph J Hauke, and Stefano Tarantolo
- Subjects
business.industry ,Regeneration (biology) ,Melanoma ,Immunology ,Cancer ,Complement C5a ,hemic and immune systems ,chemical and pharmacologic phenomena ,respiratory system ,medicine.disease ,Differential effects ,Article ,Myeloid cells ,Cancer research ,Humans ,Immunology and Allergy ,Medicine ,Myeloid Cells ,Tumor promotion ,business ,Cell Proliferation - Abstract
The specific role of C5a in cancer, especially in melanoma, has yet to be determined. Differential effects of C5a could be cancer specific. In the host defense system, C5a functions to protect the body from harmful entities via a plethora of mechanisms. Yet, C5a may also serve to potentiate cancerous process. C5a facilitates cellular proliferation and regeneration by attracting myeloid-derived suppressor cells and supporting tumor promotion. In this article, we critically reviewed the properties, mechanisms of action and functions of C5a, with particular emphasis on cancer inhibition and promotion, and clinical application of such knowledge in better management of patients with cancer. Outstanding questions and future directions in regard to the function of C5a in melanoma and other cancers are discussed.
- Published
- 2014
44. Efficacy and safety of nivolumab in patients with non-clear cell renal cell carcinoma (RCC): Results from the phase IIIb/IV CheckMate 374 study
- Author
-
Elaine T. Lam, Ana M. Molina, Joshua Zhang, Huanyu Zhao, Scott S. Tykodi, Mark D. Kochenderfer, Edward Arrowsmith, Ralph J. Hauke, Jennifer L. Johansen, Bradley G. Somer, Todd M. Bauer, Nicholas J. Vogelzang, Joshua Jemison McFarlane, Gurjyot K. Doshi, Vijay Gunuganti, and Rohit Jain
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Checkmate ,medicine.disease ,03 medical and health sciences ,Clear cell renal cell carcinoma ,Safety profile ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,In patient ,Nivolumab ,business ,030215 immunology - Abstract
562 Background: Initial safety results from the phase 3b/4 CheckMate 374 study showed that flat-dose nivolumab (NIVO) at 240 mg every 2 wk (Q2W) had a consistent safety profile across patients (pts) with clear cell and non-clear cell advanced RCC. We report updated safety and first disclosure of efficacy for pts with non-clear cell RCC (nccRCC) in CheckMate 374. Methods: Eligible pts in this cohort were adults with advanced or metastatic nccRCC who received 0–3 prior systemic therapies. Pts received NIVO 240 mg IV Q2W for ≤24 mo or until confirmed progression, unacceptable toxicity, or withdrawal of consent. Pts who benefited after 24 mo continued treatment according to the standard of care. The primary endpoint was incidence of high-grade immune-mediated adverse events (IMAEs). Exploratory endpoints included overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and duration of response (DOR). Results: In CheckMate 374, 44 pts had nccRCC. Histological subtypes included papillary (n = 24), chromophobe (n = 7), unclassified (n = 8), and other (n = 5). Most pts with nccRCC (66%) were treatment-naïve. After a median follow-up of 11.1 mo, median OS was 16.3 mo (95% confidence interval [CI] 9.2–not estimable [NE]). OS was similar regardless of baseline PD-L1 expression. ORR was 13.6% (95% CI 5.2–27.4). One pt had complete response (chromophobe histology) and 5 pts had partial response (2 pts with papillary and 1 pt each with chromophobe, collecting duct, and unclassified histology). Median DOR was 10.2 mo (95% CI 5.6–NE). Median PFS was 2.2 mo (95% CI 1.8–5.4). The 1-year PFS rate was 14% (95% CI 5–27). No new safety concerns were identified. No treatment-related grade 5 AEs or grade 3–4 IMAEs were reported. Conclusions: Clinically meaningful antitumor activity was observed in the first prospective study of NIVO monotherapy in nccRCC. Responses were observed in several histological subtypes. The safety profile of flat-dose NIVO at 240 IV Q2W is consistent with the initial outcomes reported from this study and across the NIVO program. Clinical trial information: NCT02596035.
- Published
- 2019
45. RENAVIV: A randomized phase III, double-blind, placebo-controlled study of pazopanib with or without abexinostat in patients with locally advanced or metastatic renal cell carcinoma
- Author
-
Luke T. Nordquist, Ralph J. Hauke, Pamela N. Munster, Rahul Aggarwal, and Scott Thomas
- Subjects
Cancer Research ,medicine.medical_specialty ,business.industry ,Urology ,Placebo-controlled study ,Abexinostat ,Locally advanced ,medicine.disease ,Clinical trial ,Pazopanib ,Double blind ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Oncology ,chemistry ,Renal cell carcinoma ,030220 oncology & carcinogenesis ,Medicine ,In patient ,business ,030215 immunology ,medicine.drug - Abstract
TPS681 Background: Abexinostat is a pan-histone deacetylase (HDAC) inhibitor that has shown promising activity in prior pre-clinical studies and early phase clinical trials. A phase 1b study of pazopanib plus abexinostat (Aggarwal et al. J Clin Oncol 2017) demonstrated strikingly durable responses in patients (pts) with clear cell renal cell carcinoma (RCC), including one patient with previously refractory disease with ongoing response for > 5 years’ duration. Induction of histone acetylation in peripheral blood mononuclear cells (PBMCs) was associated with durable treatment response. We hypothesize that the addition of abexinostat to pazopanib will significantly improve outcomes in patients with clear cell RCC. Methods: RENAVIV is a global, randomized, double-blind, placebo-controlled, two arm phase 3 study of pazopanib plus abexinostat versus pazopanib plus placebo, in pts with locally advanced or metastatic RCC with clear cell component. Up to one prior line of immunotherapy is allowed. Prior VEGF-targeting tyrosine kinase inhibitor treatment is prohibited. Stratification factors include: 1) Prior immunotherapy (yes/no) and 2) prognostic group (good, intermediate, poor). Pts randomized to pazopanib + placebo have the option of crossing over to receive pazopanib plus abexinostat at the time of disease progression. The primary endpoint is PFS by independent review committee. Secondary endpoints include PFS by investigator assessment, overall survival, safety, objective response rate (ORR), duration of response, patient-reported quality of life, and outcomes in cross-over population. Planned correlative studies include association between histone acetylation and HDAC expression in PBMCs with clinical outcomes. The total planned accrual is 413 pts, estimated to provide 90% power to detect a hazard ratio of 0.67 in the comparison of PFS between experimental versus control arms, with an overall two-sided type I error rate of 0.025. A pre-specified minimum of 50% of patients are required to have received prior immunotherapy. The first patient was enrolled in October 2018. Clinical trial information: NCT03592472.
- Published
- 2019
46. Non-urothelial bladder cancer: Genomic alterations and patient outcomes
- Author
-
David Arguello, Daniel M. Geynisman, Daniel A. Vaena, Matthew Zibelman, Chethan Ramamurthy, Ralph J. Hauke, Fern Anari, Earle F. Burgess, Nancy A. Dawson, Elisabeth I. Heath, Donald E. Henson, Bradley G. Somer, Benjamin Miron, Jeanny B. Aragon-Ching, Pooja Ghatalia, Thomas F. Hogan, and Elizabeth R. Plimack
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Standard of care ,Bladder cancer ,business.industry ,medicine.disease ,stomatognathic diseases ,Internal medicine ,Medicine ,Adenocarcinoma ,Basal cell ,business ,neoplasms - Abstract
399 Background: Adenocarcinoma (ADA) and squamous cell carcinoma (SCC) are rare, aggressive subtypes of bladder cancer, for which no clear standard of care exists. We report on survival of pts with ADA and SCC and identify potential therapeutic targets using molecular profiling via next generation sequencing (NGS). Methods: Survival trends, demographics, pt characteristics were obtained from the Surveillance, Epidemiology, and End Results (SEER) Database. In a separate cohort, NGS results from 72 specimens (50% metastatic) were also analyzed, using either a hotspot 47 gene panel or a 592 gene assay (Caris Life Sciences, Phoenix, AZ). Results: In SEER, 235,537 cases of bladder cancer were extracted from 1988-2008, of which 3096 were SCC and 671 were ADA. 90% of pts were white, although more African-American patients (15%) were seen in those with ADA. Among all stages, median overall survival (mOS) and 5-yr survival rates were 17.9 mos and 58% for ADA and 15 mos and 37% for SCC. Via NGS testing, 43 patients (28 ADA, 15 SCC) were tested with a 47 gene panel and 29 (21 ADA, 8 SCC) with a 592 gene panel. In the 47 gene panel, among ADA pts, the highest mutation rates were TP53 (57.1%), KRAS (21.4%), SMAD4 (14.8%), PIK3CA (10.7%) and BRCA2 (7.7%). Among SCC pts, the highest mutation rates were TP53 (66.7%), PIK3CA (33.3%), HRAS (14.3%), FBXW7 (6.7%) and AKT1 (6.7%). In the 592 gene assay, the genes with the highest mutation rates in pts with ADA were TP53 (81%), SMAD4 (33.3%), KRAS (23.8%), KMT2C (11.8%), ARID1A (11.1%), BRAF (9.5%), CTNNB1 (9.5%), KMT2D (9.5%), TSC1 (9.5%), KDM6A (5.9%), CDKN2A (5%). Among SCC pts, the highest mutation rates were TP53 (75%), CDKN2A (42.9%), FGFR3 (25%), PIK3CA (25%), CIC (14.3%), KDM6A (14.3%), BRAF (12.5%), BRCA1 (12.5%), FH (12.5%), HRAS (12.5%) and KMT2D (12.5%). Only 1 pt had high TMB. Conclusions: Genomic profiling identifies differences in underlying tumor biology of bladder ADA and SCC, which on a population level are rare with poor survival. Overall, the alterations in the PIK3CA/ AKT/ mTOR and TP53 pathways are similar to what has been reported in UC. Future analyses of these malignancies should investigate the emerging actionable targets, such as TSC1, FGFR3, BRCA1/2 and BRAF.
- Published
- 2019
47. Molecular profiling of aggressive variant urothelial carcinoma
- Author
-
Ralph J. Hauke, Nancy A. Dawson, David Arguello, Daniel A. Vaena, Daniel M. Geynisman, Elizabeth R. Plimack, Jeanny B. Aragon-Ching, Fern Anari, Thomas F. Hogan, Pooja Ghatalia, Earle F. Burgess, Chethan Ramamurthy, Matthew Zibelman, and Elisabeth I. Heath
- Subjects
03 medical and health sciences ,Cancer Research ,0302 clinical medicine ,Oncology ,business.industry ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,business ,030215 immunology ,Urothelial carcinoma - Abstract
378 Background: The WHO recognizes multiple variant histologies of urothelial carcinoma (vUC), many of which have been associated with poor outcomes compared with urothelial carcinoma (UC). We aimed to explore molecular differences between aggressive vUC and UC. Methods: 23 micropapillary (MP), 16 plasmacytoid (P), 23 sarcomatoid (S), 7 nested (N), 6 clear cell (CC), and 2 giant cell (GC) vUC specimens were tested between 2012 to 2018 via a multiplatform profiling service (Caris Life Sciences, Phoenix, AZ) consisting of gene sequencing (next generation sequencing [NGS]), gene amplification (CISH or NGS), and protein expression (immunohistochemistry [IHC]). Findings were compared to 435 control UC specimens using the Chi-square test. Results: 84% of samples were from primary tumor. Alterations identified are summarized in Table 1, and are notable for high rates of TP53 mutations across histologic subtypes, varying rates of RB1, ERBB2 and FGFR mutations, and overall low rates of DNA damage repair (DDR) mutations (29 genes reported) except in S. There were more ARID1A mutations detected in MP than UC (100% [3 specimens] v. 41.3%, p=0.044), and more CDH1 mutations in P than UC (50% [4 specimens] v. 2%, p5%) in a high proportion of S (55.6%, p=0.002) but in a lower proportion of other vUC (e.g. absent in P). Tumor mutational burden (TMB) was high in a lower proportion of vUC: 18.4% UC vs. 14.3% MP, 0% P, 16.7% S. Conclusions: Aggressive variant histology UCs have a differential profile of molecular aberrations compared to UC, with notable differences in potential targets such as HER2 and DDR genes as well as immunotherapy biomarkers. Further studies are needed to confirm these findings, and may support therapy development for these rare, aggressive UC subtypes. Aberrations (%) in Variant Histology UC. [Table: see text]
- Published
- 2019
48. Cellular Immunotherapy Study of Prostate Cancer Patients and Resulting IgG Responses to Peptide Epitopes Predicted From Prostate Tumor-associated Autoantigens
- Author
-
William Jay Ramsey, Ralph J. Hauke, Gabriela R. Rossi, Charles J. Link, Vladimir M. Pisarev, Charles A. Enke, Nicholas N. Vahanian, George P. Hemstreet, Lucinda Tennant, and Laura Helfner
- Subjects
Male ,Cancer Research ,medicine.medical_treatment ,Immunology ,Adenocarcinoma ,Autoantigens ,Cancer Vaccines ,Epitope ,Prostate cancer ,Immune system ,Antigen ,Antigens, Neoplasm ,Prostate ,Cell Line, Tumor ,medicine ,Humans ,Immunology and Allergy ,Aged ,Aged, 80 and over ,Pharmacology ,business.industry ,Immunogenicity ,Prostatic Neoplasms ,Immunotherapy ,Middle Aged ,medicine.disease ,Vaccination ,medicine.anatomical_structure ,Immunoglobulin G ,Peptides ,business ,Trisaccharides - Abstract
The immunogenicity of a cellular immunotherapy using genetically modified vaccines to express α(1,3)galactosyl epitopes (αGal) was evaluated in advanced prostate cancer (PC) patients. In this dose escalation phase I study, we report safety, feasibility, and immunologic data of an immunotherapy composed of 2 human PC cell lines engineered to express αGal epitopes (HyperAcute-Prostate, HAP, NewLink Genetics). Eight patients received up to 12 biweekly vaccinations with HAP. Enrolled patients (aged range, 53-85 y) had American Joint Committee on Cancer stage IV, any T, any N, M1, Eastern Cooperative Oncology Group PS≤2, at least 1 prior hormonal treatment and3 prior chemotherapies, adequate bone marrow and organ function, and albumin ≥3.0 g/dL. Serum IgG antibodies to synthetic peptides overexpressed in PC were determined by enzyme-linked immunosorbent assay. Results indicate that HAP immunotherapy induced humoral immune responses to autoantigens in 2 of 8 patients. These patients developed IgG antibody to multiple epitopes overexpressed in PC after immunization. These responding patients received higher doses of the immunotherapy suggesting a dose response. Two immunogenic proteins (prostate-specific membrane antigen, hepsin) belong to the extracellular molecules family participating in malignant cell invasion. Median overall survival for patients was 25.1 months with 1 patient surviving over 70 months with stable PSA and bone metastasis before expiring of other causes. Three of 8 patients showed PSA stabilization (100 d). In conclusion, HAP immunotherapy induces IgG responses to epitopes from autoantigens overexpressed in PC suggesting dose-dependent effect. HAP represents a viable immunotherapy approach to induce immune responses against tumor cells and may provide clinical benefit with minimal toxicity.
- Published
- 2013
49. MUC5AC interactions with integrin β4 enhances the migration of lung cancer cells through FAK signaling
- Author
-
Ralph J. Hauke, Surinder K. Batra, Apar Kishor Ganti, Shiv Ram Krishn, Sukhwinder Kaur, Sonny L. Johansson, Imayavaramban Lakshmanan, George B. Carey, S Paknikar, Moorthy P. Ponnusamy, Rama Krishna Nimmakayala, Parthasarathy Seshacharyulu, Saswati Karmakar, Satyanarayana Rachagani, and Garima Kaushik
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Lung Neoplasms ,Integrin ,Biology ,Mucin 5AC ,medicine.disease_cause ,digestive system ,Article ,Metastasis ,Focal adhesion ,03 medical and health sciences ,Mice ,0302 clinical medicine ,fluids and secretions ,Cell Movement ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Phosphorylation ,Lung cancer ,Molecular Biology ,Cell Proliferation ,Integrin beta4 ,Cell growth ,respiratory system ,medicine.disease ,respiratory tract diseases ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Focal Adhesion Protein-Tyrosine Kinases ,Cancer research ,biology.protein ,Adenocarcinoma ,KRAS ,sense organs ,Cisplatin ,Signal Transduction - Abstract
MUC5AC is a secretory mucin aberrantly expressed in various cancers. In lung cancer, MUC5AC is overexpressed in both primary and metastatic lesions; however, its functional role is not well understood. The present study was aimed at evaluating mechanistic role of MUC5AC on metastasis of lung cancer cells. Clinically, the overexpression of MUC5AC was observed in lung cancer patient tissues and was associated with poor survival. In addition, the overexpression of Muc5ac was also observed in genetically engineered mouse lung adenocarcinoma tissues (Kras(G12D); Trp53(R172H/+); AdCre) in comparison with normal lung tissues. Our functional studies showed that MUC5AC knockdown resulted in significantly decreased migration in two lung cancer cell lines (A549 and H1437) as compared with scramble cells. Expression of integrins (α5, β1, β3, β4 and β5) was decreased in MUC5AC knockdown cells. As both integrins and MUC5AC have a von Willebrand factor domain, we assessed for possible interaction of MUC5AC and integrins in lung cancer cells. MUC5AC strongly interacted only with integrin β4. The co-localization of MUC5AC and integrin β4 was observed both in A549 lung cancer cells as well as genetically engineered mouse adenocarcinoma tissues. Activated integrins recruit focal adhesion kinase (FAK) that mediates metastatic downstream signaling pathways. Phosphorylation of FAK (Y397) was decreased in MUC5AC knockdown cells. MUC5AC/integrin β4/FAK-mediated lung cancer cell migration was confirmed through experiments utilizing a phosphorylation (Y397)-specific FAK inhibitor. In conclusion, overexpression of MUC5AC is a poor prognostic marker in lung cancer. MUC5AC interacts with integrin β4 that mediates phosphorylation of FAK at Y397 leading to lung cancer cell migration.
- Published
- 2016
50. Sequential Therapy in Metastatic Renal Cell Carcinoma
- Author
-
Bradford R Hirsch, Thomas E. Hutson, Gury Doshi, Ralph J. Hauke, John M. Burke, Manish Agrawal, Nicholas J. Vogelzang, and Mark D. Fleming
- Subjects
medicine.medical_specialty ,renal cell carcinoma ,sequential therapy ,business.industry ,kidney cancer ,Review Article ,medicine.disease ,lcsh:Diseases of the genitourinary system. Urology ,lcsh:RC870-923 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,urologic and male genital diseases ,lcsh:RC254-282 ,Surgery ,03 medical and health sciences ,0302 clinical medicine ,Renal cell carcinoma ,030220 oncology & carcinogenesis ,Medicine ,metastasis ,030212 general & internal medicine ,business ,Intensive care medicine ,Kidney cancer - Abstract
The treatment of metastatic renal cell carcinoma (mRCC) has changed dramatically in the past decade. As the number of available agents, and related volume of research, has grown, it is increasingly complex to know how to optimally treat patients. The authors are practicing medical oncologists at the US Oncology Network, the largest community-based network of oncology providers in the country, and represent the leadership of the Network's Genitourinary Research Committee. We outline our thought process in approaching sequential therapy of mRCC and the use of real-world data to inform our approach. We also highlight the evolving literature that will impact practicing oncologists in the near future.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.