151
|
Meyo MT, Boudou-Rouquette P, Arrondeau J, Yu Chen JQ, Hirsch L, Neveux N, Fabre E, Guidet C, Damotte D, Wislez M, Alexandre J, Durand JP, Ulmann G, Goldwasser F. Relationship between energetic gap and sensitivity to anti-programmed cell death 1 immune checkpoint inhibitors in non-small cell lung cancer patients: The ELY-2 study. Clin Nutr ESPEN 2024; 64:44-50. [PMID: 39244156 DOI: 10.1016/j.clnesp.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/17/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND & AIMS We previously reported in the ELY prospective study that increased resting energy expenditure (REE) - so-called hypermetabolism - worsened tumor response, 6-month progression-free (PFS) and overall survival (OS) in metastatic non-small cell lung cancer (mNSCLC) patients treated with immune checkpoint inhibitors (ICI). Here, we investigated the effect of caloric coverage on the sensitivity to ICI. METHODS We retrospectively analysed a multicentric database of mNSCLC patients treated with ICI. All patients had a baseline nutritional assessment including REE measured with indirect calorimetry and a dietitian estimation of food intakes. Measured/theoretical REE ≥110% defined hypermetabolism. Intakes ≥90% of estimated needs defined caloric coverage. The primary endpoint was PFS. Secondary endpoints included response rate and OS. RESULTS Among 162 patients, 84 (51.9%) were normometabolic, and 78 (48.1%) hypermetabolic. In hypermetabolic patients, 40 (51.3%) met their caloric needs (group A) while 38 (48.7%) did not (group B). Median PFS was 4.3 vs. 1.9 months in groups A and B, respectively (HR: 0.49, 95%CI [0.31-0.80], p = 0.004). The PFS achieved in the group A and in normometabolic patients were similar (HR: 0.99, 95%CI [0.65-1.51], p = 0.95). In multivariate analysis, caloric coverage was independently associated with improved PFS in hypermetabolic patients (HR: 0.56, 95%CI [0.31-0.99], p = 0.048). Among hypermetabolic patients, the median OS was higher in the group A (HR: 0.58, 95%CI [0.35-0.95], p = 0.03). CONCLUSION Energy supply is a critical determinant of the sensitivity to ICI in NSCLC patients. A randomized study to evaluate the benefit of early nutritional intervention is warranted.
Collapse
Affiliation(s)
- Manuela Tiako Meyo
- Department of Medical Oncology, Cochin Hospital, AP-HP, CARPEM, France; Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France.
| | - Pascaline Boudou-Rouquette
- Department of Medical Oncology, Cochin Hospital, AP-HP, CARPEM, France; Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France
| | - Jennifer Arrondeau
- Department of Medical Oncology, Cochin Hospital, AP-HP, CARPEM, France; Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France
| | - Jeanne Qiong Yu Chen
- Department of Medical Oncology, Cochin Hospital, AP-HP, CARPEM, France; Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France
| | - Laure Hirsch
- Department of Medical Oncology, Cochin Hospital, AP-HP, CARPEM, France; Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France
| | - Nathalie Neveux
- Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France; Department of Medical Biology, Cochin Hospital, AP-HP, Paris, France
| | - Elizabeth Fabre
- Department of Thoracic Oncology, Georges Pompidou European Hospital, AP-HP, CARPEM, France; INSERM U970, Université Paris Cité, INSERM, PARCC, F-75015 Paris, France
| | - Caroline Guidet
- Department of Medical Oncology, Cochin Hospital, AP-HP, CARPEM, France; Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France
| | - Diane Damotte
- Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France; Paris Cité University, France; Department of Pathology, Cochin Hospital, APHP, Paris, France
| | - Marie Wislez
- Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France; Paris Cité University, France; Department of Pneumology, Thoracic Oncology Unit, Institut du Cancer Paris Carpem, Cochin Hospital, APHP, Paris, France
| | - Jérôme Alexandre
- Department of Medical Oncology, Cochin Hospital, AP-HP, CARPEM, France; Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France; Paris Cité University, France; Institut des Cordeliers, INSERM U1147, Paris, France
| | - Jean-Philippe Durand
- Department of Medical Oncology, Cochin Hospital, AP-HP, CARPEM, France; Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France
| | - Guillaume Ulmann
- Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France; Department of Medical Biology, Cochin Hospital, AP-HP, Paris, France
| | - François Goldwasser
- Department of Medical Oncology, Cochin Hospital, AP-HP, CARPEM, France; Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France; Paris Cité University, France; Institut Cochin, INSERM U1016, Paris, France
| |
Collapse
|
152
|
Cánovas-Cervera I, Nacher-Sendra E, Suay G, Lahoz A, García-Giménez JL, Mena-Mollá S. Role of miRNAs as epigenetic regulators of immune checkpoints in lung cancer immunity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 390:109-139. [PMID: 39864893 DOI: 10.1016/bs.ircmb.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The advent of immunotherapy in cancer has provided new avenues in the treatment of many malignancies at various stages. Specifically, immune checkpoint inhibitors (ICIs) have transformed the field of lung cancer treatment. However, since some tumors can evade the immune system, not all patients respond properly. Recent research has provided evidence showing how microRNAs (miRNAs) are involved in regulating many immune checkpoints. MiRNAs have demonstrated their ability to modulate immune evasion of tumor cells. Currently, reliable markers are being sought to predict the efficacy of immunotherapy in these types of cancers. Therefore, the association of serum miRNAs and the response of ICIs in lung cancer is under study. Many miRNA molecules and their corresponding target genes have been identified in the regulation of chemoresistance. Therefore, elucidating how these miRNAs control the function of immune checkpoints, as well as the effectiveness of therapies based on ICIs set the basis for the development of new biomarkers to predict treatment response to ICIs. This chapter delves into the molecular role of miRNAs interacting with ICs, such as PD-1 and PD-L1, and the clinical utility of miRNAs, such as miR-16, miR-146a, and miR-335, in predicting treatment response to ICI-based therapy in lung cancer. The aim is to provide a deep insight of the current landscape, serving as a cornerstone for further research.
Collapse
Affiliation(s)
- Irene Cánovas-Cervera
- INCLIVA Health Research Institute, INCLIVA, Valencia, Spain; Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - Elena Nacher-Sendra
- INCLIVA Health Research Institute, INCLIVA, Valencia, Spain; Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - Guillermo Suay
- Medical Oncology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Agustin Lahoz
- Biomarkers and Precision Medicine Unit, Health Research Institute-Hospital La Fe, Valencia, Spain; Analytical Unit, Health Research Institute-Hospital La Fe, Valencia, Spain
| | - José Luis García-Giménez
- INCLIVA Health Research Institute, INCLIVA, Valencia, Spain; Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain; Consortium Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Valencia, Spain.
| | - Salvador Mena-Mollá
- INCLIVA Health Research Institute, INCLIVA, Valencia, Spain; Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjassot, Spain
| |
Collapse
|
153
|
Hokamura N, Fukagawa T, Fukushima R, Kiyokawa T, Horikawa M, Soeda N, Suzuki Y, Kaneshiro S, Abe K, Kodashima S, Yamamoto T, Oshima Y, Ishida T, Sasajima Y, Nomoto A, Shiraishi K, Ito A. Evaluation of pembrolizumab plus cisplatin and fluorouracil in radical treatment for patients with T4b esophageal squamous cell carcinoma. BMC Gastroenterol 2024; 24:295. [PMID: 39223478 PMCID: PMC11370125 DOI: 10.1186/s12876-024-03382-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Pembrolizumab plus cisplatin and 5-fluorouracil administered as first-line therapy for advanced esophageal cancer patients has shown a better objective response and survival than conventional chemotherapy with less severe hematological adverse events. The safety and efficacy of this regimen were evaluated in patients with T4b esophageal squamous cell carcinoma (ESCC). METHODS Eight consecutive patients with T4b ESCC received this regimen according to KEYNOTE-590 as induction, and they were evaluated after 1-3 courses. The programmed death-ligand 1 (PD-L1) combined positive score (CPS) was also evaluated before chemotherapy. Efficacy for the primary lesion was evaluated by our original formula for the tumor reduction rate. RESULTS The numbers of patients with partial response (PR), stable disease, and progressive disease (PD) were 5, 1, and 2, respectively. The tumor reduction rate ranged from 69 to 87% in PR patients, and all PR patients had relief from T4b. Two patients underwent conversion surgery with R0 resection. PD-L1 CPS was over 90 in 2 PR patients, but under 10 in 2 other PR patients. PD-L1 CPS was under 10 in PD patients. One patient had hyperprogression, resulting in an esophago-pulmonary fistula. Greater than grade 3 adverse events were bleeding gastric ulcer in one patient (12.5%), neutropenia without G-CSF in 3 patients (37.5%), and hypopotassemia in 1 patient (12.5%). No patient had febrile neutropenia. CONCLUSIONS Marked tumor reduction was confirmed in 62.5% of patients with pembrolizumab plus cisplatin and 5-fluorouracil with less adverse events. This regimen could be administered as induction chemotherapy for patients with T4b ESCC.
Collapse
Affiliation(s)
- Nobukazu Hokamura
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan.
| | - Takeo Fukagawa
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Ryoji Fukushima
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Takashi Kiyokawa
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Masahiro Horikawa
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Naruyoshi Soeda
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Yusuke Suzuki
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Shinya Kaneshiro
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Koichiro Abe
- Department of Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Shinya Kodashima
- Department of Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Takatsugu Yamamoto
- Department of Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Yasutoshi Oshima
- Department of Pathology, Teikyo University School of Medicine, Tokyo, Japan
| | - Tsuyoshi Ishida
- Department of Pathology, Teikyo University School of Medicine, Tokyo, Japan
| | - Yuko Sasajima
- Department of Pathology, Teikyo University School of Medicine, Tokyo, Japan
| | - Akihiro Nomoto
- Department of Radiology, Teikyo University School of Medicine, Tokyo, Japan
| | - Kenshiro Shiraishi
- Department of Radiology, Teikyo University School of Medicine, Tokyo, Japan
| | - Ai Ito
- Department of Pharmacy, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
154
|
Schroeder T, Martens T, Fransecky L, Valerius T, Schub N, Pott C, Baldus C, Stölzel F. Management of chimeric antigen receptor T (CAR-T) cell-associated toxicities. Intensive Care Med 2024; 50:1459-1469. [PMID: 39172238 PMCID: PMC11377606 DOI: 10.1007/s00134-024-07576-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/21/2024] [Indexed: 08/23/2024]
Abstract
The use of chimeric antigen receptor T (CAR-T) cells is a significant therapeutic improvement increasing the prognosis for patients with a variety of hematological malignancies. However, this therapy has also sometimes life-threatening, complications. Therefore, knowledge of the treatment and management of these complications, especially in treatment centers and intensive care units, respectively, is of outstanding importance. This review provides recommendations for the diagnosis, management, and treatment of CAR-T cell-associated complications such as cytokine release syndrome, immune effector cell associated neurotoxicity syndrome, hematotoxicity, hypogammaglobulinemia, and CAR-T cell-induced pseudo-progression amongst others for physicians treating patients with CAR-T cell-associated complications and intensivists.
Collapse
Affiliation(s)
- Torsten Schroeder
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Tjark Martens
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Lars Fransecky
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Thomas Valerius
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Natalie Schub
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Christiane Pott
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Claudia Baldus
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Friedrich Stölzel
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany.
| |
Collapse
|
155
|
Wagner S, Ewald C, Freitag D, Herrmann KH, Koch A, Bauer J, Vogl TJ, Kemmling A, Gufler H. Radiomics and visual analysis for predicting success of transplantation of heterotopic glioblastoma in mice with MRI. J Neurooncol 2024; 169:257-267. [PMID: 38960965 PMCID: PMC11341603 DOI: 10.1007/s11060-024-04725-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 05/25/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Quantifying tumor growth and treatment response noninvasively poses a challenge to all experimental tumor models. The aim of our study was, to assess the value of quantitative and visual examination and radiomic feature analysis of high-resolution MR images of heterotopic glioblastoma xenografts in mice to determine tumor cell proliferation (TCP). METHODS Human glioblastoma cells were injected subcutaneously into both flanks of immunodeficient mice and followed up on a 3 T MR scanner. Volumes and signal intensities were calculated. Visual assessment of the internal tumor structure was based on a scoring system. Radiomic feature analysis was performed using MaZda software. The results were correlated with histopathology and immunochemistry. RESULTS 21 tumors in 14 animals were analyzed. The volumes of xenografts with high TCP (H-TCP) increased, whereas those with low TCP (L-TCP) or no TCP (N-TCP) continued to decrease over time (p < 0.05). A low intensity rim (rim sign) on unenhanced T1-weighted images provided the highest diagnostic accuracy at visual analysis for assessing H-TCP (p < 0.05). Applying radiomic feature analysis, wavelet transform parameters were best for distinguishing between H-TCP and L-TCP / N-TCP (p < 0.05). CONCLUSION Visual and radiomic feature analysis of the internal structure of heterotopically implanted glioblastomas provide reproducible and quantifiable results to predict the success of transplantation.
Collapse
Affiliation(s)
- Sabine Wagner
- Department of Neuroradiology, Marburg University Hospital - Philipps University, 35043, Marburg, Germany.
- Department of Neuroradiology, Institute for Diagnostic and Interventional Radiology, Jena University Hospital - Friedrich Schiller University, 07747, Jena, Germany.
| | - Christian Ewald
- Department of Neurosurgery, Brandenburg Medical School, Theodor Fontane, University Hospital Brandenburg/Havel, 14770, Brandenburg/Havel, Germany
| | - Diana Freitag
- Department of Neurosurgery, Section of Experimental Neurooncology, Jena University Hospital - Friedrich Schiller University, 07747, Jena, Germany
| | - Karl-Heinz Herrmann
- Medical Physics Group, Institute for Diagnostic and Interventional Radiology, Jena University Hospital - Friedrich Schiller University, 07743, Jena, Germany
| | - Arend Koch
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, and Berlin Institute of Health, Charité University Medicine, 10117, Berlin, Germany
| | - Johannes Bauer
- Department of Neurosurgery, Brandenburg Medical School, Theodor Fontane, University Hospital Brandenburg/Havel, 14770, Brandenburg/Havel, Germany
| | - Thomas J Vogl
- Department of Diagnostic and Interventional Radiology, Goethe University Hospital Frankfurt, 60590, Frankfurt Am Main, Germany
| | - André Kemmling
- Department of Neuroradiology, Marburg University Hospital - Philipps University, 35043, Marburg, Germany
| | - Hubert Gufler
- Department of Diagnostic and Interventional Radiology, Goethe University Hospital Frankfurt, 60590, Frankfurt Am Main, Germany
| |
Collapse
|
156
|
Bessudo A, Haseeb AM, Reeves JA, Zhu X, Wong L, Giranda V, Suttner L, Liu F, Chatterjee M, Sharma S. Safety and Efficacy of Vicriviroc (MK-7690) in Combination With Pembrolizumab in Patients With Advanced or Metastatic Microsatellite Stable Colorectal Cancer. Clin Colorectal Cancer 2024; 23:285-294. [PMID: 38942693 DOI: 10.1016/j.clcc.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Pembrolizumab, a monoclonal antibody against PD-1, has shown limited efficacy in patients with microsatellite stable or mismatch repair proficient (MSS/pMMR) metastatic colorectal cancer (CRC). We evaluated vicriviroc (small-molecule C-C motif chemokine ligand 5 antagonist) plus pembrolizumab in patients with advanced or metastatic MSS/pMMR CRC. PATIENTS AND METHODS This open-label, phase 2 trial (NCT03631407) enrolled adults with histologically confirmed, locally advanced, unresectable or metastatic CRC that was MSS per local assessment. All patients had received previous treatment with standard therapies. Patients were randomized 1:1 to vicriviroc 150 mg orally once daily plus pembrolizumab 200 mg intravenously every 3 weeks or vicriviroc 250 mg orally once daily plus pembrolizumab 200 mg intravenously every 3 weeks for up to 35 cycles (2 years). Primary endpoints were the objective response rate (ORR) as assessed by the investigator per RECIST v1.1, dose-limiting toxicities (DLTs), adverse events (AEs), and discontinuations due to AEs. RESULTS Forty patients were enrolled and treated. ORR was 5% (95% CI, 0.1%-24.9%) in both treatment groups. There were no complete responses; 1 patient in each treatment group experienced a partial response. No patient in the vicriviroc 150 mg plus pembrolizumab group experienced a DLT. Two patients in the vicriviroc 250 mg plus pembrolizumab group experienced DLTs (1 grade 4 encephalopathy and 1 grade 4 pneumonitis). CONCLUSION The combination of vicriviroc at doses of 150 or 250 mg plus pembrolizumab 200 mg showed limited antitumor activity in patients with advanced or metastatic MSS/pMMR CRC. Toxicity with the combination was manageable.
Collapse
Affiliation(s)
- Alberto Bessudo
- California Cancer Associates for Research and Excellence, Encinitas, CA
| | | | - James A Reeves
- Florida Cancer Specialists and Research Institute/Sarah Cannon Research Institute, Fort Myers, FL
| | - Xiaofu Zhu
- Cross Cancer Institute, Edmonton, AB, Canada
| | - Lucas Wong
- Baylor College of Medicine, Houston, TX; Baylor Scott and White Health, Vasicek Cancer Treatment Center, Temple, TX
| | | | | | | | | | | |
Collapse
|
157
|
Young RWC, Rodriguez GR, Kucera J, Carrera D, Antevil JL, Trachiotis GD. Molecular Markers, Immune Therapy, and Non-Small Cell Lung Cancer-State-of-the-Art Review for Surgeons. J Laparoendosc Adv Surg Tech A 2024; 34:786-797. [PMID: 38900703 DOI: 10.1089/lap.2024.0164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024] Open
Abstract
Background: Lung cancer is a leading cause of cancer deaths in the United States. An increasing understanding of relevant non-small cell lung cancer (NSCLC) biomarkers has led to the recent development of molecular-targeted therapies and immune checkpoint inhibitors that have revolutionized treatment for patients with advanced and metastatic disease. The purpose of this review is to provide surgeons with a state-of-the-art understanding of the current medical and surgical treatment trends and their implications in the future of management of NSCLC. Materials and Methods: A systematic search of PubMed was conducted to identify English language articles published between January 2010 and March 2024 focusing on molecular markers, tumor targeting, and immunotherapy in the diagnosis and treatment of NSCLC. Case series, observational studies, randomized trials, guidelines, narrative reviews, systematic reviews, and meta-analyses were included. Results: There is now increasing data to suggest that molecular-targeted therapies and immune therapies have a role in the neoadjuvant setting. Advances in intraoperative imaging allow surgeons to perform increasingly parenchymal-sparing lung resections without compromising tumor margins. Liquid biopsies can noninvasively detect targetable mutations in cancer cells and DNA from a blood draw, potentially allowing for earlier diagnosis, personalized therapy, and long-term monitoring for disease recurrence. Conclusions: The management of NSCLC has advanced dramatically in recent years fueled by a growing understanding of the cancer biology of NSCLC. Advances in medical therapies, surgical techniques, and diagnostic and surveillance modalities continue to evolve but have already impacted current treatment strategies for NSCLC, which are encompassed in this review.
Collapse
Affiliation(s)
- Robert W C Young
- Department of Surgery, George Washington University Hospital, Washington, District of Columbia, USA
| | - Gustavo R Rodriguez
- Department of Surgery, George Washington University Hospital, Washington, District of Columbia, USA
| | - John Kucera
- Department of Surgery, Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Daniel Carrera
- Department of Surgery, George Washington University Hospital, Washington, District of Columbia, USA
| | - Jared L Antevil
- Department of Surgery, George Washington University Hospital, Washington, District of Columbia, USA
- Division of Cardiothoracic Surgery and Heart Center, Washington DC Veterans Affairs Medical Center, Washington, District of Columbia, USA
| | - Gregory D Trachiotis
- Department of Surgery, George Washington University Hospital, Washington, District of Columbia, USA
- Division of Cardiothoracic Surgery and Heart Center, Washington DC Veterans Affairs Medical Center, Washington, District of Columbia, USA
| |
Collapse
|
158
|
Hassler MR, Moedlagl V, Hindinger H, Krauter J, Klager S, Resch I, Huebner N, Yurdakul O, Ofner H, Korn SM, D'Andrea D, Gust K, Shariat SF. Treatment Patterns and Real-World Outcomes for Locally Advanced or Metastatic Urothelial Cancer in the Era of Immunotherapy. Eur Urol Focus 2024; 10:779-787. [PMID: 38161107 DOI: 10.1016/j.euf.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/07/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND AND OBJECTIVE There are limited data on real-world outcomes for patients with advanced or metastatic urothelial cancer (mUC) since immune checkpoint inhibitors (ICIs) became available. Our objective was to analyze outcomes for patients with mUC since ICIs became available. METHODS We performed a retrospective analysis of 131 patients with mUC attending the outpatient clinic of a single tertiary care center who received systemic therapy between June 2017 and July 2021 with follow-up up to December 2022. Summary and descriptive statistics were calculated for categorical and continuous variables. The Kaplan-Meier method was applied to calculate survival, and a Cox proportional-hazards model was used to explore associations between clinical variables and outcomes. KEY FINDINGS AND LIMITATIONS The median patient age was 68 yr (range 35-90). The first systemic therapy administered was platinum-based in 79% of cases and ICI-based in 21%. Some 61% of the cohort received a second systemic treatment, with 75% of these an ICI. Median overall survival for the entire cohort was 24 mo (interquartile range 9-35). Patients on ICI therapy for ≥6 mo had median overall survival of 59 mo (95% confidence interval 39 mo-not reached). Metastatic sites on initiation of ICI therapy and C-reactive protein kinetics were prognostic in patients receiving ICIs. Limitations include the retrospective design and inherent selection bias. CONCLUSIONS AND CLINICAL IMPLICATIONS More than 60% of patients with mUC received second-line treatment, and 75% of these received an ICI. Patients staying on immunotherapy for more than 6 mo have substantially better outcomes in comparison to patients with less time on immunotherapy and historical cohorts. PATIENT SUMMARY We looked at the lines of therapy and outcomes for patients with advanced or metastatic cancer of the urinary tract, starting from when immunotherapy drugs called immune checkpoint inhibitors (ICIs) became available. We found that 60% of patients have received second-line therapy, which is a double the rate in comparison to historical groups of patients. Patients with long-term ICI therapy (>6 months) had significantly better outcomes, with a median survival of more than 3 years.
Collapse
Affiliation(s)
- Melanie R Hassler
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Verena Moedlagl
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Hanna Hindinger
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Johanna Krauter
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Sonja Klager
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Irene Resch
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Nicolai Huebner
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Ozan Yurdakul
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Heidemarie Ofner
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Stephan M Korn
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - David D'Andrea
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Kilian Gust
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Shahrokh F Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Weill Cornell Medical College, New York, NY, USA; Department of Urology, University of Texas Southwestern, Dallas, TX, USA; Department of Urology, Second Faculty of Medicine, Charles University, Prague, Czechia; Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan; Karl Landsteiner Institute of Urology and Andrology, Vienna, Austria.
| |
Collapse
|
159
|
Colombo N, Biagioli E, Harano K, Galli F, Hudson E, Antill Y, Choi CH, Rabaglio M, Marmé F, Marth C, Parma G, Fariñas-Madrid L, Nishio S, Allan K, Lee YC, Piovano E, Pardo B, Nakagawa S, McQueen J, Zamagni C, Manso L, Takehara K, Tasca G, Ferrero A, Tognon G, Lissoni AA, Petrella M, Laudani ME, Rulli E, Uggeri S, Barretina Ginesta MP. Atezolizumab and chemotherapy for advanced or recurrent endometrial cancer (AtTEnd): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2024; 25:1135-1146. [PMID: 39102832 DOI: 10.1016/s1470-2045(24)00334-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND At the time of AtTEnd trial design, standard treatment for advanced or recurrent endometrial cancer included carboplatin and paclitaxel chemotherapy. This trial assessed whether combining atezolizumab with chemotherapy might improve outcomes in this population. METHODS AtTEnd was a multicentre, double-blind, randomised, placebo-controlled, phase 3 trial done in 89 hospitals in 11 countries across Europe, Australia, New Zealand, and Asia. Enrolled patients were aged 18 years or older, and had advanced or recurrent endometrial carcinoma or carcinosarcoma, an Eastern Cooperative Oncology Group performance status of 0-2, and received no previous systemic chemotherapy for recurrence. Patients were randomly assigned (2:1) using an interactive web response system (block size of six) to either atezolizumab 1200 mg or placebo given intravenously with chemotherapy (carboplatin at area under the curve of 5 or 6 and paclitaxel 175 mg/m2 intravenously on day 1 every 21 days) for 6-8 cycles, then continued until progression. Stratification factors were country, histological subtype, advanced or recurrent status, and mismatch repair (MMR) status. Participants and treating clinicians were masked to group allocation. The hierarchically tested co-primary endpoints were progression-free survival (in patients with MMR-deficient [dMMR] tumours, and in the overall population) and overall survival (in the overall population). Primary analyses were done in the intention-to-treat population, defined as all randomly assigned patients who gave their full consent to participation in the study and data processing. Safety was assessed in all patients included in the intention-to-treat population who received at least one dose of study treatment. Here, we report the primary progression-free survival and the interim overall survival results. This study is ongoing and is registered with ClinicalTrials.gov, NCT03603184. FINDINGS Between Oct 3, 2018, and Jan 7, 2022, 551 patients were randomly assigned to atezolizumab (n=362) or placebo (n=189). Two patients in the atezolizumab group were excluded from all analyses due to lack of consent. Median follow-up was 28·3 months (IQR 21·2-37·6). 81 (23%) patients in the atezolizumab group and 44 (23%) patients in the placebo group had dMMR disease by central assessment. In the dMMR population, median progression-free survival was not estimable (95% CI 12·4 months-not estimable [NE]) in the atezolizumab group and 6·9 months (6·3-10·1) in the placebo group (hazard ratio [HR] 0·36, 95% CI 0·23-0·57; p=0·0005). In the overall population, median progression-free survival was 10·1 months (95% CI 9·5-12·3) in the atezolizumab group and 8·9 months (8·1-9·6) in the placebo group (HR 0·74, 95% CI 0·61-0·91; p=0·022). Median overall survival was 38·7 months (95% CI 30·6-NE) in the atezolizumab group and 30·2 months (25·0-37·2) in the placebo group (HR 0·82, 95% CI 0·63-1·07; log-rank p=0·048). The p value for the interim analysis of overall survival did not cross the stopping boundary; therefore, the trial will continue until the required number of events are recorded. The most common grade 3-4 adverse events were neutropenia (97 [27%] of 356 patients in the atezolizumab group vs 51 [28%] of 185 in the placebo group) and anaemia (49 [14%] vs 24 [13%]). Treatment-related serious adverse events occurred in 46 (13%) patients in the atezolizumab group and six (3%) patients in the placebo group. Treatment-related deaths occurred in two patients (pneumonia in one patient in each group). INTERPRETATION Atezolizumab plus chemotherapy increased progression-free survival in patients with advanced or recurrent endometrial carcinoma, particularly in those with dMMR carcinomas, suggesting the addition of atezolizumab to standard chemotherapy as first-line treatment in this specific subgroup. FUNDING F Hoffmann-La Roche.
Collapse
Affiliation(s)
- Nicoletta Colombo
- European Institute of Oncology IRCCS, Milan, Italy; University of Milan-Bicocca, Milan, Italy
| | - Elena Biagioli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| | - Kenichi Harano
- Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Francesca Galli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Yoland Antill
- Peninsula Health, Monash University, Melbourne, VIC, Australia
| | - Chel Hun Choi
- Department of Obstetrics and Gynecology, Samsung Seoul Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Manuela Rabaglio
- Department of Medical Oncology, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Frederic Marmé
- Obstetrics and Gynecology, Section Gynecologic Oncology, Universitaetsklinikum Mannheim Medizinische Fakultaet, Mannheim, Germany
| | - Christian Marth
- Department of Obstetrics and Gynecology, Medical University Innsbruck, Innsbruck, Austria
| | | | | | - Shin Nishio
- Department of Obstetrics and Gynecology, Kurume University School of Medicine, Kurume, Japan
| | - Karen Allan
- Glasgow Oncology Clinical Trials Unit, University of Glasgow, Glasgow, UK
| | - Yeh Chen Lee
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Elisa Piovano
- AOU Città della Salute e della Scienza di Torino - Presidio Sant'Anna, Torino, Italy
| | - Beatriz Pardo
- Department of Medical Oncology, Catalan Institute of Oncology, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Satoshi Nakagawa
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - John McQueen
- Glasgow Oncology Clinical Trials Unit, University of Glasgow, Glasgow, UK
| | - Claudio Zamagni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Kazuhiro Takehara
- Department of Gynecology, NHO Shikoku Cancer Center, Matsuyama, Japan
| | - Giulia Tasca
- UOC Oncologia 2 IRCCS Istituto Oncologico Veneto - Padova, Padova, Italy
| | - Annamaria Ferrero
- Academic Division Obstetrics and Gynaecology, Mauriziano Hospital, University Department of Surgical Sciences, Torino, Italy
| | - Germana Tognon
- UO Ostetricia-Ginecologia ASST Spedali Civili, Università degli Studi-Brescia, Brescia, Italy
| | - Andrea Alberto Lissoni
- UOS Oncologia Ginecologica Medica, Università degli Studi di Milano Bicocca, IRCCS San Gerardo dei Tintori - Monza, Monza, Italy
| | - Mariacristina Petrella
- SOD Oncologia Medica Ginecologica, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Maria Elena Laudani
- AOU Città della Salute e della Scienza di Torino - Presidio Sant'Anna, Torino, Italy
| | - Eliana Rulli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Sara Uggeri
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | |
Collapse
|
160
|
O'Neal HR, Sheybani R, Kraus CK, Self WH, Shah AM, Thomas CB, Tse HTK, Scoggins R. Cellular host response sepsis test for risk stratification of patients in the emergency department: A pooled analysis. Acad Emerg Med 2024; 31:883-893. [PMID: 38643433 DOI: 10.1111/acem.14923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/06/2024] [Accepted: 03/31/2024] [Indexed: 04/22/2024]
Abstract
OBJECTIVES Sepsis is one of the most common, costly, and misdiagnosed conditions in U.S. emergency departments (EDs). ED providers often treat on nonspecific signs, subjective suspicion, or presumption of infection, resulting in over- and undertreatment. An increased understanding of host response has opened a new direction for sepsis diagnostics. The IntelliSep test is a U.S. Food and Drug Administration-cleared cellular host response diagnostic that could help distinguish sepsis in ED settings. Our objective was to evaluate the potential of the cellular host response test to expedite appropriate care for patients who present with signs of infection. METHODS We performed a pooled analysis of five adult (≥18 years) cohorts enrolled at seven geographically diverse U.S. sites in separate studies. Structured blinded adjudication was used to classify presence or absence of sepsis, and only patients with high confidence in the adjudicated label were included (n = 1002), defined as patients for whom there was consensus in the determination of sepsis per the Sepsis-3 and severe sepsis per the Sepsis-2 definitions between both the independent adjudication panel and the site-level physician. RESULTS Among patients with signs or suspicion of infection, the test achieved similar or better performance compared to other indicators in identifying patients at high risk for sepsis (specificity > 83%) and significantly superior performance in identifying those at low risk (sensitivity > 92%; 0% sepsis-associated mortality). The test also stratified severity of illness, as shown by 30-day in-hospital mortality (p < 0.001), hospital length of stay (p < 0.01), and use of hospital resources (p < 0.001). CONCLUSIONS Our data suggest that the cellular host response test provides clinically actionable results for patients at both high and low risk for sepsis and provides a rapid, objective means for risk stratification of patients with signs of infection. If integrated into standard of care, the test may help improve outcomes and reduce unnecessary antibiotic use.
Collapse
Affiliation(s)
- Hollis R O'Neal
- Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Our Lady of the Lake Regional Medical Center, Baton Rouge, Louisiana, USA
| | | | - Chadd K Kraus
- Department of Emergency and Hospital Medicine, Lehigh Valley Health Network, Allentown, Pennsylvania, USA
- University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Wesley H Self
- Department of Emergency Medicine, Vanderbilt Institute for Clinical and Translational Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Emergency Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ajay M Shah
- Cytovale, Inc., San Francisco, California, USA
| | - Christopher B Thomas
- Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Franciscan Missionaries of Our Lady Health System, Baton Rouge, Louisiana, USA
| | | | - Robert Scoggins
- Cytovale, Inc., San Francisco, California, USA
- Pulmonary & Critical Care, Kootenai Health, Coeur d'Alene, Idaho, USA
| |
Collapse
|
161
|
Men Q, Duan Y, Pei F, Yao Q, He W, Zhao Y, Shi L, Liu G, Huang J. PD-1 blockade combined with chemotherapy and bevacizumab in DNA mismatch repair-proficient/microsatellite stable colorectal liver metastases. J Gastrointest Oncol 2024; 15:1534-1544. [PMID: 39279968 PMCID: PMC11399864 DOI: 10.21037/jgo-23-940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 07/03/2024] [Indexed: 09/18/2024] Open
Abstract
Background Single-agent immunotherapy is less effective in patients with DNA mismatch repair-proficient/microsatellite stable (pMMR/MSS) metastatic colorectal cancer (mCRC). Whether pMMR/MSS mCRC patients benefit from combination immunotherapy remains unclear. This study aimed to evaluate the efficacy and safety of anti-programmed cell death protein 1 (PD-1) therapy combined with chemotherapy and bevacizumab in pMMR/MSS colorectal liver metastases (CRLM) patients. Methods A total of 12 patients with pMMR/MSS CRLM treated at The Sixth Affiliated Hospital of Sun Yat-sen University were enrolled. All patients were treated with at least 4 doses of PD-1 monoclonal antibody combined with chemotherapy and bevacizumab as neoadjuvant/adjuvant therapy. Results A total of 10 of the 12 patients received the combined therapies before primary tumor resection; the disease control rate (DCR) was 100% (10/10), and the objective response rate (ORR) was 70% (7/10). The ORR of liver metastases was 75% (9/12). Pathological complete response (pCR) was achieved in 1 primary tumor patient and 2 patients with hepatic lesions. A total of 5 patients underwent simultaneous resection of the primary tumor and liver metastases; 9 patients underwent microwave ablation for liver metastases. A total of 7 patients were assessed as having no evidence of disease (NED) with a median progression-free survival (PFS) interval of 9.2 (1.5-15.8) months after multimodality treatments for both primary and metastatic lesions. No severe immune-related adverse events (irAEs) and operational complications were observed. Conclusions PD-1 blockade combined with chemotherapy and bevacizumab might be safe and effective for patients with pMMR/MSS CRLM. This treatment strategy might lead to better tumor regression and a higher chance of achieving NED.
Collapse
Affiliation(s)
- Qianqian Men
- Graceland Medical Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yinghua Duan
- Department of Traditional Chinese Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fengyun Pei
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qijun Yao
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wan He
- Department of Oncology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Yandong Zhao
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lishuo Shi
- Clinical Research Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guangjian Liu
- Department of Medical Ultrasonic, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun Huang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangzhou, China
| |
Collapse
|
162
|
Cao LM, Zhong NN, Chen Y, Li ZZ, Wang GR, Xiao Y, Liu XH, Jia J, Liu B, Bu LL. Less is more: Exploring neoadjuvant immunotherapy as a de-escalation strategy in head and neck squamous cell carcinoma treatment. Cancer Lett 2024; 598:217095. [PMID: 38964728 DOI: 10.1016/j.canlet.2024.217095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/15/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) constitutes a significant global cancer burden, given its high prevalence and associated mortality. Despite substantial progress in survival rates due to the enhanced multidisciplinary approach to treatment, these methods often lead to severe tissue damage, compromised function, and potential toxicity. Thus, there is an imperative need for novel, effective, and minimally damaging treatment modalities. Neoadjuvant treatment, an emerging therapeutic strategy, is designed to reduce tumor size and curtail distant metastasis prior to definitive intervention. Currently, neoadjuvant chemotherapy (NACT) has optimized the treatment approach for a subset of HNSCC patients, yet it has not produced a noticeable enhancement in overall survival (OS). In the contemporary cancer therapeutics landscape, immunotherapy is gaining traction at an accelerated pace. Notably, neoadjuvant immunotherapy (NAIT) has shown promising radiological and pathological responses, coupled with encouraging efficacy in several clinical trials. This potentially paves the way for a myriad of possibilities in treatment de-escalation of HNSCC, which warrants further exploration. This paper reviews the existing strategies and efficacies of neoadjuvant immune checkpoint inhibitors (ICIs), along with potential de-escalation strategies. Furthermore, the challenges encountered in the context of the de-escalation strategies of NAIT are explored. The aim is to inform future research directions that strive to improve the quality of life (QoL) for patients battling HNSCC.
Collapse
Affiliation(s)
- Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Somatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Somatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Somatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Somatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Somatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Somatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Xuan-Hao Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Somatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Jun Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Somatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Somatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Somatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
163
|
Marković F, Stjepanović M, Samardžić N, Kontić M. The Association of Immune-Related Adverse Events with the Efficacy of Atezolizumab in Previously Treated Advanced Non-Small-Cell Lung Cancer Patients: A Single-Center Experience. Cancers (Basel) 2024; 16:2995. [PMID: 39272852 PMCID: PMC11394052 DOI: 10.3390/cancers16172995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) are pivotal in managing metastatic non-oncogene addicted non-small-cell lung cancer (NSCLC). They have unique toxicities known as immune-related adverse events (irAEs). Previous studies have linked irAEs during atezolizumab-based first-line treatments in advanced NSCLC with improved outcomes. This study explored the association between irAEs and the efficacy of atezolizumab in advanced NSCLC patients who had previously received platinum-based chemotherapy. The study involved 105 advanced NSCLC patients who received atezolizumab monotherapy after progressing on at least one line of platinum-based chemotherapy from a single academic institution in Serbia. Data were obtained from a hospital lung cancer registry. Among the participants, 63.8% were male, with the majority being current (53.3%) or former smokers (37.1%). About half had a good performance status (ECOG PS 0-1) at the start of atezolizumab treatment. irAEs occurred in 23 patients (21.9%). The median progression-free survival (mPFS) was significantly longer for patients with irAEs (13.03 months) compared to those without (3.4 months) (HR 0.365 [95% CI, 0.195-0.681], p = 0.002). irAEs and ECOG PS 0-1 were predictors of longer mPFS, with irAEs being more common in patients with good performance status (p = 0.01). irAEs were linked to improved mPFS in NSCLC patients treated with atezolizumab after multiple lines of platinum-based chemotherapy.
Collapse
Affiliation(s)
- Filip Marković
- Clinic for Pulmonology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Mihailo Stjepanović
- Clinic for Pulmonology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Natalija Samardžić
- Clinic for Pulmonology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Milica Kontić
- Clinic for Pulmonology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
164
|
Patel RK, Parappilly MS, Walker BS, Heussner RT, Fung A, Chang YH, Kardosh A, Lopez CD, Mayo SC, Wong MH. Exploratory Analyses of Circulating Neoplastic-Immune Hybrid Cells as Prognostic Biomarkers in Advanced Intrahepatic Cholangiocarcinoma. Int J Mol Sci 2024; 25:9198. [PMID: 39273147 PMCID: PMC11395231 DOI: 10.3390/ijms25179198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
Existing clinical biomarkers do not reliably predict treatment response or disease progression in patients with advanced intrahepatic cholangiocarcinoma (ICC). Circulating neoplastic-immune hybrid cells (CHCs) have great promise as a blood-based biomarker for patients with advanced ICC. Peripheral blood specimens were longitudinally collected from patients with advanced ICC enrolled in the HELIX-1 phase II clinical trial (NCT04251715). CHCs were identified by co-expression of pan-cytokeratin (CK) and CD45, and levels were correlated to patient clinical disease course. Unsupervised machine learning was then performed to extract their morphological features to compare them across disease courses. Five patients were included in this study, with a median of nine specimens collected per patient. A median of 13.5 CHCs per 50,000 peripheral blood mononuclear cells were identified at baseline, and levels decreased to zero following the initiation of treatment in all patients. Counts remained undetectable in three patients who demonstrated end-of-trial clinical treatment response and conversely increased in two patients with evidence of therapeutic resistance. In the post-trial surveillance period, interval counts increased prior to or at the time of clinical progression in three patients and remain undetectable in one patient with continued long-term disease stability. Using our machine learning platform, treatment-resistant CHCs exhibited upregulation of CK and downregulation of CD45 relative to treatment-responsive CHCs. CHCs represent a promising blood-based biomarker to supplement traditional radiographic and biochemical measures.
Collapse
Affiliation(s)
- Ranish K. Patel
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA; (R.K.P.)
| | - Michael S. Parappilly
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
| | - Brett S. Walker
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA; (R.K.P.)
| | - Robert T. Heussner
- Department of Biomedical Engineering, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
| | - Alice Fung
- Department of Diagnostic Radiology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
| | - Adel Kardosh
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
- Department of Medicine, Division of Medical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| | - Charles D. Lopez
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
- Department of Medicine, Division of Medical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| | - Skye C. Mayo
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA; (R.K.P.)
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
| | - Melissa H. Wong
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
| |
Collapse
|
165
|
Wang Y, Sun C, Liu M, Xu P, Li Y, Zhang Y, Huang J. Dysregulated gene expression of SUMO machinery components induces the resistance to anti-PD-1 immunotherapy in lung cancer by upregulating the death of peripheral blood lymphocytes. Front Immunol 2024; 15:1424393. [PMID: 39211047 PMCID: PMC11357960 DOI: 10.3389/fimmu.2024.1424393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Background The majority of patients with lung cancer exhibit drug resistance after anti-PD-1 immunotherapy, leading to shortened patient survival time. Previous studies have suggested an association between epigenetic abnormalities such as methylation and clinical response to anti-PD-1 immunotherapy, while the role of SUMOylation in resistance to anti-PD-1 antibody immunotherapy is still unclear. Methods Here, the mRNA expression of 15 SUMO machinery components in PBMC from lung cancer patients receiving anti-PD-1 immunotherapy were analyzed using real-time PCR. Base on the percentage change in mRNA levels, the relationship between the expression of SUMO machinery components and outcomes of anti-PD-1 immunotherapy, and the influencing factors of SUMOylation were evaluated. PBMC was treated with different concentrations of 2-D08 (a specific inhibitor of SUMOylation) in vitro, and analyzed the activation and the death rates of lymphocyte subsets by flow cytometry analysis. Results A predictive method, base on the gene expression of three SUMO machinery components (SUMO1, SUMO3 and UBE2I), were developed to distinguish non-responders to PD-1 inhibitors. Furthermore, the number of lymphocytes in peripheral blood significantly reduced in the dysregulated SUMOylation groups (the percentage change >100 or -50 ~ -100 groups). In vitro studies confirmed that lightly low SUMOylation level improved the activation status of T and NK lymphocytes, but extremely low SUMOylation level lead to the increased death rates of lymphocytes. Conclusion Our findings implied that dysregulated gene expression of SUMO machinery components could induce the resistance of anti-PD-1 immunotherapy in lung cancer by upregulating the death of peripheral blood lymphocytes. These data might provide effective circulating biomarkers for predicting the efficacy of anti-PD-1 immunotherapy, and uncovered a novel regulatory mechanism of resistance to anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Ying Wang
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Chao Sun
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Mengmeng Liu
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Panyang Xu
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yanyan Li
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yongsheng Zhang
- Prenatal Diagnosis Center, Reproductive Medicine Center, The First Hospital of Jilin University, Changchun, China
| | - Jing Huang
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
166
|
Butner JD, Dogra P, Chung C, Koay EJ, Welsh JW, Hong DS, Cristini V, Wang Z. Hybridizing mechanistic modeling and deep learning for personalized survival prediction after immune checkpoint inhibitor immunotherapy. NPJ Syst Biol Appl 2024; 10:88. [PMID: 39143136 PMCID: PMC11324794 DOI: 10.1038/s41540-024-00415-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/29/2024] [Indexed: 08/16/2024] Open
Abstract
We present a study where predictive mechanistic modeling is combined with deep learning methods to predict individual patient survival probabilities under immune checkpoint inhibitor (ICI) immunotherapy. This hybrid approach enables prediction based on both measures that are calculable from mechanistic models of key mechanisms underlying ICI therapy that may not be directly measurable in the clinic and easily measurable quantities or patient characteristics that are not always readily incorporated into predictive mechanistic models. A deep learning time-to-event predictive model trained on a hybrid mechanistic + clinical data set from 93 patients achieved higher per-patient predictive accuracy based on event-time concordance, Brier score, and negative binomial log-likelihood-based criteria than when trained on only mechanistic model-derived values or only clinical data. Feature importance analysis revealed that both clinical and model-derived parameters play prominent roles in increasing prediction accuracy, further supporting the advantage of our hybrid approach.
Collapse
Affiliation(s)
- Joseph D Butner
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- The Cameron School of Business, University of St. Thomas, Houston, TX, USA.
| | - Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eugene J Koay
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Medical Education, Texas A&M University School of Medicine, Bryan, TX, USA.
| |
Collapse
|
167
|
Andersen MB, Drljevic-Nielsen A, Ehlers JH, Thorup KS, Baandrup AO, Palne M, Rasmussen F. DCE-CT parameters as new functional imaging biomarkers at baseline and during immune checkpoint inhibitor therapy in patients with lung cancer - a feasibility study. Cancer Imaging 2024; 24:105. [PMID: 39135095 PMCID: PMC11320886 DOI: 10.1186/s40644-024-00745-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 07/24/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND With the development of immune checkpoint inhibitors for the treatment of non-small cell lung cancer, the need for new functional imaging techniques and early response assessments has increased to account for new response patterns and the high cost of treatment. The present study was designed to assess the prognostic impact of dynamic contrast-enhanced computed tomography (DCE-CT) on survival outcomes in non-small cell lung cancer patients treated with immune checkpoint inhibitors. METHODS Thirty-three patients with inoperable non-small-cell lung cancer treated with immune checkpoint inhibitors were prospectively enrolled for DCE-CT as part of their follow-up. A single target lesion at baseline and subsequent follow-up examinations were enclosed in the DCE-CT. Blood volume deconvolution (BVdecon), blood flow deconvolution (BFdecon), blood flow maximum slope (BFMax slope) and permeability were assessed using overall survival (OS) and progression-free survival (PFS) as endpoints in Kaplan Meier and Cox regression analyses. RESULTS High baseline Blood Volume (BVdecon) (> 12.97 ml × 100 g-1) was associated with a favorable OS (26.7 vs 7.9 months; p = 0.050) and PFS (14.6 vs 2.5 months; p = 0.050). At early follow-up on day seven a higher relative increase in BFdecon (> 24.50% for OS and > 12.04% for PFS) was associated with an unfavorable OS (8.7 months vs 23.1 months; p < 0.025) and PFS (2.5 vs 13.7 months; p < 0.018). The relative change in BFdecon (categorical) on day seven was a predictor of OS (HR 0.26, CI95: 0.06 to 0.93 p = 0.039) and PFS (HR 0.27, CI95: 0.09 to 0.85 p = 0.026). CONCLUSION DCE-CT-identified parameters may serve as potential prognostic biomarkers at baseline and during early treatment in patients with NSCLC treated with immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Michael Brun Andersen
- Department of Radiology, Copenhagen University Hospital, Gentofte, Denmark.
- Department of Radiology, Zealand University Hospital, Køge, Denmark.
- Department of Radiology, Aarhus University Hospital, Skejby, Denmark.
- Radiology Department, Copenhagen University Hospital, Gentofte Hospitalsvej 1, 2900, Hellerup, Denmark.
- Department of clinical medicine, Copenhagen University, Copenhagen, Denmark.
| | | | | | | | | | - Majbritt Palne
- Department of Radiology, Zealand University Hospital, Køge, Denmark
| | - Finn Rasmussen
- Department of Radiology, Aarhus University Hospital, Skejby, Denmark
| |
Collapse
|
168
|
Orillard E, Adhikari A, Malouf RS, Calais F, Marchal C, Westeel V. Immune checkpoint inhibitors plus platinum-based chemotherapy compared to platinum-based chemotherapy with or without bevacizumab for first-line treatment of older people with advanced non-small cell lung cancer. Cochrane Database Syst Rev 2024; 8:CD015495. [PMID: 39136258 PMCID: PMC11320659 DOI: 10.1002/14651858.cd015495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
BACKGROUND Lung cancer is a cancer of the elderly, with a median age at diagnosis of 71. More than one-third of people diagnosed with lung cancer are over 75 years old. Immune checkpoint inhibitors (ICIs) are special antibodies that target a pathway in the immune system called the programmed cell death 1/programmed cell death-ligand 1 (PD-1/PD-L1) pathway. These antibodies help the immune system fight cancer cells by blocking signals that cancer cells use to avoid being attacked by the immune system. ICIs have changed the treatment of people with lung cancer. In particular, for people with previously-untreated advanced non-small cell lung cancer (NSCLC), current first-line treatment now comprises ICIs plus platinum-based chemotherapy, rather than platinum-based chemotherapy alone, regardless of their PD-L1 expression status. However, as people age, their immune system changes, becoming less effective in its T cell responses. This raises questions about how well ICIs work in older adults. OBJECTIVES To assess the effects of immune checkpoint inhibitors (ICIs) in combination with platinum-based chemotherapy compared to platinum-based chemotherapy (with or without bevacizumab) in treatment-naïve adults aged 65 years and older with advanced NSCLC. SEARCH METHODS We searched the Cochrane Lung Cancer Group Trial Register, CENTRAL, MEDLINE, Embase, two other trial registers, and the websites of drug regulators. The latest search date was 23 August 2023. We also checked references and searched abstracts from the meetings of seven cancer organisations from 2019 to August 2023. SELECTION CRITERIA We included randomised controlled trials (RCTs) that reported on the efficacy and safety of adding ICIs to platinum-based chemotherapy compared to platinum-based chemotherapy alone for people 65 years and older who had not previously been treated. All data emanated from international multicentre studies involving adults with histologically-confirmed advanced NSCLC who had not received any previous systemic anticancer therapy for their advanced disease. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. Our primary outcomes were overall survival and treatment-related adverse events (grade 3 or higher). Our secondary outcomes were progression-free survival, objective response rate, time to response, duration of response, and health-related quality of life (HRQoL). MAIN RESULTS We included 17 primary studies, with a total of 4276 participants, in the review synthesis. We identified nine ongoing studies, and listed one study as 'awaiting classification'. Twelve of the 17 studies included people older than 75 years, accounting for 9% to 13% of their participants. We rated some studies as having 'some concerns' for risk of bias arising from the randomisation process, deviations from the intended interventions, or measurement of the outcome. The overall GRADE rating for the certainty of the evidence ranged from moderate to low because of the risk of bias, imprecision, or inconsistency. People aged 65 years and older The addition of ICIs to platinum-based chemotherapy probably increased overall survival compared to platinum-based chemotherapy alone (hazard ratio (HR) 0.78, 95% confidence interval (CI) 0.70 to 0.88; 8 studies, 2093 participants; moderate-certainty evidence). Only one study reported data for treatment-related adverse events (grade 3 or higher). The frequency of treatment-related adverse events may not differ between the two treatment groups (risk ratio (RR) 1.09, 95% CI 0.89 to 1.32; 1 study, 127 participants; low-certainty evidence). The addition of ICIs to platinum-based chemotherapy probably improves progression-free survival (HR 0.61, 95% CI 0.54 to 0.68; 7 studies, 1885 participants; moderate-certainty evidence). People aged 65 to 75 years, inclusive The addition of ICIs to platinum-based chemotherapy probably improved overall survival compared to platinum-based chemotherapy alone (HR 0.75, 95% CI 0.65 to 0.87; 6 studies, 1406 participants; moderate-certainty evidence). Only one study reported data for treatment-related adverse events (grade 3 or higher). The frequency of treatment-related adverse events probably increased in people treated with ICIs plus platinum-based chemotherapy compared to those treated with platinum-based chemotherapy alone (RR 1.47, 95% CI 1.02 to 2.13; 1 study, 97 participants; moderate-certainty evidence). The addition of ICIs to platinum-based chemotherapy probably improved progression-free survival (HR 0.64, 95% CI 0.57 to 0.73; 8 studies, 1466 participants; moderate-certainty evidence). People aged 75 years and older There may be no difference in overall survival in people treated with ICIs combined with platinum-based chemotherapy compared to platinum-based chemotherapy alone (HR 0.90, 95% CI 0.70 to 1.16; 4 studies, 297 participants; low-certainty evidence). No data on treatment-related adverse events were available in this age group. The effect of combination ICI and platinum-based chemotherapy on progression-free survival is uncertain (HR 0.83, 95% CI 0.51 to 1.36; 3 studies, 226 participants; very low-certainty evidence). Only three studies assessed the objective response rate. For time to response, duration of response, and health-related quality of life, we do not have any evidence yet. AUTHORS' CONCLUSIONS Compared to platinum-based chemotherapy alone, adding ICIs to platinum-based chemotherapy probably leads to higher overall survival and progression-free survival, without an increase in treatment-related adverse events (grade 3 or higher), in people 65 years and older with advanced NSCLC. These data are based on results from studies dominated by participants between 65 and 75 years old. However, the analysis also suggests that the improvements reported in overall survival and progression-free survival may not be seen in people older than 75 years.
Collapse
Affiliation(s)
- Emeline Orillard
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
- EFS, INSERM, UMR RIGHT, Université de Franche-Comté, CHU Besançon, Besançon, France
| | - Arjab Adhikari
- Internal Medicine, Ascension Saint Francis Hospital, Evanston, Illinois, USA
| | - Reem S Malouf
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - François Calais
- Bibliothèque Universitaire de Santé, Université de Franche-Comté, Besançon, France
| | | | - Virginie Westeel
- EFS, INSERM, UMR RIGHT, Université de Franche-Comté, CHU Besançon, Besançon, France
- Department of Chest Diseases and Thoracic Oncology, University Hospital of Besançon, Besançon, France
| |
Collapse
|
169
|
Beulque Y, Kinget L, Roussel E, Mobaraki S, Laenen A, Debruyne PR, Van Herck Y, Baldewijns M, Wozniak A, Garg AD, Zucman-Rossi J, Couchy G, Albersen M, De Wever L, Haaker L, Beuselinck B. Baseline neutrophil-to-eosinophil-ratio and outcome in metastatic clear-cell renal cell carcinoma treated with nivolumab or ipilimumab/nivolumab. Acta Oncol 2024; 63:658-668. [PMID: 39129249 PMCID: PMC11332468 DOI: 10.2340/1651-226x.2024.40390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/28/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND AND PURPOSE This study aims to evaluate neutrophil-to-eosinophil ratio (NER) as a prognostic and/or predictive biomarker in metastatic clear cell renal cell carcinoma (m-ccRCC) treated with nivolumab or ipilimumab/nivolumab. PATIENTS/MATERIALS AND METHODS We performed a retrospective study on m-ccRCC patients treated with nivolumab or ipilimumab/nivolumab (2012-2022). Baseline NER was calculated and correlated with clinical outcomes: response rate (RR), progression free survival (PFS) and overall survival (OS). Corresponding transcriptomic data were analysed. RESULTS We included 201 m-ccRCC patients, 76 treated with ipilimumab/nivolumab and 125 with nivolumab. Baseline NER was statistically significantly associated with International Metastatic RCC Database Consortium (IMDC) risk groups. Increased NER was associated with shorter PFS and OS in the total patient series and nivolumab-treated patients. In patients treated with ipilimumab/nivolumab, increased NER was only statistically significantly associated with shorter OS. The impact of baseline NER on PFS and OS was independent of IMDC risk stratification. No clear correlation was found between baseline NER and RECIST response or maximal tumour shrinkage. In two additional databases, NER was also associated with PFS and OS in first-line vascular-endothelial-growth-factor-receptor tyrosine-kinase-inhibitors (VEGFR-TKIs), but not to disease-free survival in the post-nephrectomy setting. Lower NER was associated with intratumoural molecular features possibly associated with better outcome on immune checkpoint inhibitors. INTERPRETATION Lower baseline NER is associated with better PFS and OS, independent of IMDC risk score, in m-ccRCC patients treated with ipilimumab/nivolumab or nivolumab. It correlates with intratumoural molecular features possibly associated with better outcome on immune checkpoint inhibitors. The predictive power of this biomarker is probably limited and insufficient for patient selection.
Collapse
Affiliation(s)
- Yana Beulque
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Lisa Kinget
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Eduard Roussel
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Sajedeh Mobaraki
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | | | - Philip R Debruyne
- Department of General Medical Oncology, AZ Groeninge, Kortrijk, Belgium; Medical Technology Research Centre (MTRC), School of Life Sciences, Anglia Ruskin University, Cambridge, UK; School of Nursing and Midwifery, University of Plymouth, Plymouth, UK
| | - Yannick Van Herck
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | | | | | - Abhishek D Garg
- Laboratory of Cell Stress & Immunity (CSI), Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jessica Zucman-Rossi
- Inserm, UMRS-1138, Génomique fonctionnelle des tumeurs solides, Centre de recherche des Cordeliers, Paris, France
| | - Gabrielle Couchy
- Inserm, UMRS-1138, Génomique fonctionnelle des tumeurs solides, Centre de recherche des Cordeliers, Paris, France
| | - Maarten Albersen
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Liesbeth De Wever
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | - Lorenz Haaker
- Department of Medical Oncology, AZ Sint Lucas, Brugge, Belgium
| | - Benoit Beuselinck
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
170
|
Roussot N, Thibaudin M, Fumet JD, Daumoine S, Hampe L, Rébé C, Limagne E, Lagrange A, Herreros V, Lecuelle J, Mananet H, Ilie A, Rageot D, Boidot R, Goussot V, Comte A, Jacob P, Beltjens F, Bergeron A, Charon-Barra C, Arnould L, Derangère V, Ladoire S, Truntzer C, Ghiringhelli F. Case report: Immune response characterization of a pseudoprogression in a PD-L1-negative, TMB-low, KEAP1/STK11 co-mutated metastatic NSCLC. Front Immunol 2024; 15:1437961. [PMID: 39170614 PMCID: PMC11335479 DOI: 10.3389/fimmu.2024.1437961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024] Open
Abstract
A patient with a PD-L1-negative, TMB-low, KEAP1/STK11 co-mutated metastatic non-small cell lung cancer (NSCLC) experienced a multisite radiological progression at 3 months after initiation of chemoimmunotherapy as first-line treatment for metastatic disease. After the radiological progression, while she was not undergoing treatment, the patient had spontaneous lesions shrinkage and further achieved a prolonged complete response. Genomic and transcriptomic data collected at baseline and at the time of pseudoprogression allowed us to biologically characterize this rare response pattern. We observed the presence of a tumor-specific T-cell response against tumor-specific neoantigens (TNAs). Endogenous retroviruses (ERVs) expression following chemoimmunotherapy was also observed, concurrent with biological features of an anti-viral-like innate immune response with type I IFN signaling and production of CXCR3-associated chemokines. This is the first biological characterization of a NSCLC pseudoprogression under chemoimmunotherapy followed by a prolonged complete response in a PD-L1-negative, TMB-low, KEAP1/STK11 co-mutated NSCLC. These clinical and biological data underline that even patients with multiple factors of resistance to immune checkpoint inhibitors could trigger a tumor-specific immune response to tumor neoantigen, leading to complete eradication of the tumor and probably a vaccinal immune response.
Collapse
Affiliation(s)
- Nicolas Roussot
- Unité Formation Recherche (UFR) des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - Marion Thibaudin
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | - Jean-David Fumet
- Unité Formation Recherche (UFR) des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - Susy Daumoine
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | - Léa Hampe
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | - Cédric Rébé
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | - Emeric Limagne
- Unité Formation Recherche (UFR) des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | - Aurélie Lagrange
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - Victor Herreros
- Department of Interventional Radiology, Centre Georges-François Leclerc, Dijon, France
| | - Julie Lecuelle
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | - Hugo Mananet
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | - Alis Ilie
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | - David Rageot
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | - Romain Boidot
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | - Vincent Goussot
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
| | - Anthony Comte
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
| | - Pierre Jacob
- Unité Formation Recherche (UFR) des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - Françoise Beltjens
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
| | - Anthony Bergeron
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
| | - Céline Charon-Barra
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
| | - Laurent Arnould
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
| | - Valentin Derangère
- Unité Formation Recherche (UFR) des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | - Sylvain Ladoire
- Unité Formation Recherche (UFR) des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - Caroline Truntzer
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | - François Ghiringhelli
- Unité Formation Recherche (UFR) des Sciences de Santé, Université Bourgogne Franche-Comté, Dijon, France
- Cancer Biology Transfer Platform, Department of Biology and Pathology of Tumors, Georges-François Leclerc Anticancer Center, UNICANCER, Dijon, France
- Equipe Therapies and Immune Response in Cancers (TIRECS), Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| |
Collapse
|
171
|
O’Hare P, Cooney T, de Blank P, Gutmann DH, Kieran M, Milde T, Fangusaro J, Fisher M, Avula S, Packer R, Fukuoka K, Mankad K, Mueller S, Waanders AJ, Opocher E, Bouffet E, Raabe E, Werle NE, Azizi AA, Robison NJ, Hernáiz Driever P, Russo M, Schouten N, van Tilburg CM, Sehested A, Grill J, Bandopadhayay P, Kilday JP, Witt O, Ashley DM, Ertl-Wagner BB, Tabori U, Hargrave DR. Resistance, rebound, and recurrence regrowth patterns in pediatric low-grade glioma treated by MAPK inhibition: A modified Delphi approach to build international consensus-based definitions-International Pediatric Low-Grade Glioma Coalition. Neuro Oncol 2024; 26:1357-1366. [PMID: 38743009 PMCID: PMC11300023 DOI: 10.1093/neuonc/noae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024] Open
Abstract
Pediatric low-grade glioma (pLGG) is the most common childhood brain tumor group. The natural history, when curative resection is not possible, is one of a chronic disease with periods of tumor stability and episodes of tumor progression. While there is a high overall survival rate, many patients experience significant and potentially lifelong morbidities. The majority of pLGGs have an underlying activation of the RAS/MAPK pathway due to mutational events, leading to the use of molecularly targeted therapies in clinical trials, with recent regulatory approval for the combination of BRAF and MEK inhibition for BRAFV600E mutated pLGG. Despite encouraging activity, tumor regrowth can occur during therapy due to drug resistance, off treatment as tumor recurrence, or as reported in some patients as a rapid rebound growth within 3 months of discontinuing targeted therapy. Definitions of these patterns of regrowth have not been well described in pLGG. For this reason, the International Pediatric Low-Grade Glioma Coalition, a global group of physicians and scientists, formed the Resistance, Rebound, and Recurrence (R3) working group to study resistance, rebound, and recurrence. A modified Delphi approach was undertaken to produce consensus-based definitions and recommendations for regrowth patterns in pLGG with specific reference to targeted therapies.
Collapse
Affiliation(s)
- Patricia O’Hare
- Department of Paediatric Oncology, Royal Belfast Hospital for Sick Children, Northern Ireland, UK
| | - Tabitha Cooney
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Broad Institute, Cambridge, Massachusetts, USA
- Day One Biopharmaceuticals, Boston, Massachusetts, USA
| | - Peter de Blank
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Broad Institute, Cambridge, Massachusetts, USA
- University of Cincinnati College of Medicine and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mark Kieran
- Day One Biopharmaceuticals, Boston, Massachusetts, USA
| | - Till Milde
- Clinical Pediatric Oncology, Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Jason Fangusaro
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael Fisher
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Shivaram Avula
- Department of Radiology, Alder Hey Children’s NHS Foundation Trust, Liverpool, UK
| | - Roger Packer
- Brain Tumor Institute, Center for Neuroscience and Behavioral Medicine, Children’s National Hospital, Washington, District of Columbia, USA
| | - Kohei Fukuoka
- Department of Hematology/Oncology, Saitama Children’s Medical Center, Saitama, Japan
| | - Kshitij Mankad
- Department of Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, Department of Radiology, London, UK
| | - Sabine Mueller
- Department of Neurology, Neurosurgery and Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Angela J Waanders
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Enrico Opocher
- Paediatric Haematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
| | - Eric Bouffet
- The Hospital for Sick Children and Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Eric Raabe
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Natacha Entz Werle
- Pediatric Onco-Hematology Department, University Hospital of Strasbourg. UMR CNRS 7021, University of Strasbourg, Strasbourg, France
| | - Amedeo A Azizi
- Department of Pediatrics and Adolescent Medicine and Comprehensive Centre of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Nathan J Robison
- Division of Hematology & Oncology, Children’s Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Pablo Hernáiz Driever
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, German HIT-LOGGIC-Registry for LGG in children and adolescents, Department of Pediatric Oncology/Hematology, Berlin, Germany
| | - Mark Russo
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | - Netteke Schouten
- Princess Maxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Cornelis M van Tilburg
- Clinical Pediatric Oncology, Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Astrid Sehested
- Department of Paediatrics and Adolescent Medicine, The University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Jacques Grill
- Department of Pediatric and Adolescent Oncology, Villejuif, France
| | - Pratiti Bandopadhayay
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Broad Institute, Cambridge, Massachusetts, USA
| | - John-Paul Kilday
- The Centre for Paediatric, Teenage and Young Adult Cancer, Institute of Cancer Sciences, University of Manchester, and Royal Manchester Children’s Hospital, Manchester, UK
| | - Olaf Witt
- Clinical Pediatric Oncology, Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - David M Ashley
- Department of Neurosurgery, The Preston Robert Tisch Brain Tumor Center. Pediatric Neuro-Oncology, Preuss Laboratory for Brain Tumor Research, Durham, North Carolina, USA
| | | | - Uri Tabori
- The Hospital for Sick Children and Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Darren R Hargrave
- UCL Great Ormond Street Institute of Child Health, Great Ormond Street Hospital for Children, London, UK
| |
Collapse
|
172
|
Sasaki Y, Matsumoto K, Takaki A, Adachi T, Takahara M, Ozato K, Takeuchi Y, Sue M, Miyake N, Wada N, Onishi H, Shiraha H, Oda T, Tsutsumi K, Nouso K, Kariyama K, Hagihara H, Moriya A, Otsuka M. Anti-PD-1 Autoantibody Predicts Survival of Patients With Hepatocellular Carcinoma Receiving Atezolizumab/Bevacizumab. GASTRO HEP ADVANCES 2024; 3:1138-1147. [PMID: 39559295 PMCID: PMC11570718 DOI: 10.1016/j.gastha.2024.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 07/26/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND AND AIMS Methods for predicting therapeutic response to immune checkpoint inhibitors in cancer therapy are in high demand. In patients with advanced hepatocellular carcinoma (HCC), atezolizumab (anti-programmed cell death-ligand 1 [PD-L1]) and bevacizumab (anti-vascular endothelial growth factor) combination therapy (Atezo/Bev therapy) is a first-line treatment. However, no reliable biomarkers are currently available to predict its efficacy. Here, we examined serum anti-PD-1 autoantibody levels as candidate biomarkers. METHODS We prospectively enrolled 63 patients with advanced HCC who received Atezo/Bev therapy. Serum anti-PD-1 autoantibody levels were measured before treatment using an indirect enzyme-linked immunosorbent assay. The correlation between the titers and response to therapy was statistically examined. RESULTS Serum anti-PD-1 autoantibody levels were not significantly associated with the treatment response in any patient. However, when examining only patients who received the Atezo/Bev as their first-line therapy, higher anti-PD-1 autoantibody levels were significantly associated with worse overall survival rates. The titer was an independent risk factor for poor prognosis (odds ratio [OR] = 7.8, P = .013), in addition to a higher neutrophil-to-lymphocyte ratio (OR = 7.1, P = .009) and lower albumin levels (OR = 14.2, P = .003). CONCLUSION Serum anti-PD-1 autoantibody levels correlated with the overall survival rate in patients who received Atezo/Bev as first-line therapy. Serum anti-PD-1 autoantibody levels may serve as new biomarkers for predicting the efficacy of immune checkpoint inhibitors in patients with HCC.
Collapse
Affiliation(s)
- Yuki Sasaki
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuyuki Matsumoto
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Akinobu Takaki
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takuya Adachi
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masahiro Takahara
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Keita Ozato
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yasuto Takeuchi
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masahiko Sue
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Nozomi Miyake
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Nozomu Wada
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hideki Onishi
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hidenori Shiraha
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takashi Oda
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Koichiro Tsutsumi
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuhiro Nouso
- Department of Gastroenterology, Okayama City Hospital, Okayama, Japan
| | - Kazuya Kariyama
- Department of Gastroenterology, Okayama City Hospital, Okayama, Japan
| | - Hiroaki Hagihara
- Department of Gastroenterology, Sumitomo Besshi Hospital, Niihama, Ehime, Japan
| | - Akio Moriya
- Department of Gastroenterology, Mitoyo General Hospital, Kanonji, Kagawa, Japan
| | - Motoyuki Otsuka
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
173
|
Liu FR, Wei XL, Feng WN, Zhao HY, Zhang Y, Wang ZQ, Zhang DS, Wang FH, Yang S, Pan W, Tian X, Men L, Wang H, Liang E, Wang C, Yang D, Zhai Y, Qiu MZ, Xu RH. Inhibitor of apoptosis proteins (IAP) inhibitor APG-1387 monotherapy or in combination with programmed cell death 1 (PD-1) inhibitor toripalimab in patients with advanced solid tumors: results from two phase I trials. ESMO Open 2024; 9:103651. [PMID: 39059062 PMCID: PMC11338093 DOI: 10.1016/j.esmoop.2024.103651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND APG-1387 is a novel second mitochondrial-derived activator of caspases mimetic, small-molecule inhibitor targeting inhibitor of apoptosis proteins. We report results from two phase I trials evaluating the tolerability, safety, and antitumor activity of APG-1387 monotherapy and APG-1387 plus toripalimab [a programmed cell death 1 (PD-1) inhibitor] for advanced solid tumors. PATIENTS AND METHODS Participants aged ≥18 years who had histologically confirmed advanced solid tumors with no appropriate standard of care (or refractory to standard care) were eligible. Patients received escalating intravenous doses of APG-1387 alone or combined with fixed-dose toripalimab (240 mg every 3 weeks) in a '3 + 3' design. Primary endpoints were dose-limiting toxicities (DLTs) and maximum tolerated dose (MTD) in the monotherapy trial, and recommended phase II dose (RP2D) in the combination therapy trial. Secondary endpoints included the pharmacokinetic and pharmacodynamic profiles and preliminary efficacy in both trials. RESULTS In the monotherapy trial, 28 subjects were enrolled and received ≥1 treatment cycle. No DLT was reported among the 28 subjects, and the MTD was not reached. One participant (3.6%) had a grade ≥3 treatment-related adverse event (TRAE) of alanine aminotransferase elevation. In efficacy analysis of 23 participants, none achieved an objective response, and the disease control rate was 21.7%. In the combination trial, 22 subjects were enrolled and included in all analyses. There was one DLT of grade 3 lipase elevation. The MTD was not reached. Four grade ≥3 TRAEs occurred in three participants (13.6%), with the most common being lipase elevation (n = 2). The RP2D was 45 mg weekly. The objective response rate was 13.6%, with complete response achieved in one subject, and the disease control rate was 54.5%. CONCLUSIONS APG-1387 45 mg weekly plus toripalimab was well tolerated and is recommended for further study, with preliminary clinical activity observed in study participants with advanced solid tumors.
Collapse
Affiliation(s)
- F-R Liu
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou
| | - X-L Wei
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou; Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou
| | - W-N Feng
- Department of Pulmonary Oncology, The First People's Hospital of Foshan, Foshan
| | - H-Y Zhao
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou
| | - Y Zhang
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou
| | - Z-Q Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou; Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou
| | - D-S Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou; Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou
| | - F-H Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou; Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou
| | - S Yang
- Department of Pulmonary Oncology, The First People's Hospital of Foshan, Foshan
| | - W Pan
- Ascentage Pharma (Suzhou) Co., Ltd., Suzhou, China
| | - X Tian
- Ascentage Pharma (Suzhou) Co., Ltd., Suzhou, China
| | - L Men
- Ascentage Pharma (Suzhou) Co., Ltd., Suzhou, China
| | - H Wang
- Ascentage Pharma (Suzhou) Co., Ltd., Suzhou, China
| | - E Liang
- Ascentage Pharma Group Inc., Rockville, USA
| | - C Wang
- Ascentage Pharma Group Inc., Rockville, USA
| | - D Yang
- Ascentage Pharma (Suzhou) Co., Ltd., Suzhou, China; Department of Experimental Research, State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Y Zhai
- Ascentage Pharma (Suzhou) Co., Ltd., Suzhou, China; Ascentage Pharma Group Inc., Rockville, USA.
| | - M-Z Qiu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou; Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou.
| | - R-H Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou; Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou.
| |
Collapse
|
174
|
Kotecha RR, Doshi SD, Knezevic A, Chaim J, Chen Y, Jacobi R, Zucker M, Reznik E, McHugh D, Shah NJ, Feld E, Aggen DH, Rafelson W, Xiao H, Carlo MI, Feldman DR, Lee CH, Motzer RJ, Voss MH. A Phase 2 Trial of Talazoparib and Avelumab in Genomically Defined Metastatic Kidney Cancer. Eur Urol Oncol 2024; 7:804-811. [PMID: 37945488 PMCID: PMC11074239 DOI: 10.1016/j.euo.2023.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 08/07/2023] [Accepted: 10/20/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Although different kidney cancers represent a heterogeneous group of malignancies, multiple subtypes including Von Hippel-Lindau (VHL)-altered clear cell renal cell carcinoma (ccRCC), fumarate hydratase (FH)- and succinate dehydrogenase (SDH)-deficient renal cell carcinoma (RCC), and renal medullary carcinoma (RMC) are affected by genomic instability. Synthetic lethality with poly ADP-ribose polymerase inhibitors (PARPis) has been suggested in preclinical models of these subtypes, and paired PARPis with immune checkpoint blockade (ICB) may achieve additive and/or synergistic effects in patients with previously treated advanced kidney cancers. OBJECTIVE To evaluate combined PARPi + ICB in treatment-refractory metastatic kidney cancer. DESIGN, SETTING, AND PARTICIPANTS We conducted a single-center, investigator-initiated phase 2 trial in two genomically selected advanced kidney cancer cohorts: (1) VHL-altered RCC with at least one prior ICB agent and one vascular endothelial growth factor (VEGF) inhibitor, and (2) FH- or SDH-deficient RCC with at least one prior ICB agent or VEGF inhibitor and RMC with at least one prior line of chemotherapy. INTERVENTION Patients received talazoparib 1 mg daily plus avelumab 800 mg intravenously every 14 d in 28-d cycles. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The primary endpoint was objective response rate (ORR) by Immune Response Evaluation Criteria in Solid Tumors at 4 mo, and the secondary endpoints included progression-free survival (PFS), overall survival, and safety. RESULTS AND LIMITATIONS Cohort 1 consisted of ten patients with VHL-altered ccRCC. All patients had previously received ICB. The ORR was 0/9 patients; one patient was not evaluable due to missed doses. In this cohort, seven patients achieved stable disease (SD) as the best response. The median PFS was 3.5 mo (95% confidence interval [CI] 1.0, 3.9 mo). Cohort 2 consisted of eight patients; four had FH-deficient RCC, one had SDH-deficient RCC, and three had RMC. In this cohort, six patients had previously received ICB. The ORR was 0/8 patients; two patients achieved SD as the best response and the median PFS was 1.2 mo (95% CI 0.4, 2.9 mo). The most common treatment-related adverse events of all grades were fatigue (61%), anemia (28%), nausea (22%), and headache (22%). There were seven grade 3-4 and no grade 5 events. CONCLUSIONS The first clinical study of combination PARPi and ICB therapy in advanced kidney cancer did not show clinical benefit in multiple genomically defined metastatic RCC cohorts or RMC. PATIENT SUMMARY We conducted a study to look at the effect of two medications, talazoparib and avelumab, in patients with metastatic kidney cancer who had disease progression on standard treatment. Talazoparib blocks the normal activity of molecules called poly ADP-ribose polymerase, which then prevents tumor cells from repairing themselves and growing, while avelumab helps the immune system recognize and kill cancer cells. We found that the combination of these agents was safe but not effective in specific types of kidney cancer.
Collapse
Affiliation(s)
- Ritesh R Kotecha
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| | - Sahil D Doshi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrea Knezevic
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joshua Chaim
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yingbei Chen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rachel Jacobi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark Zucker
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ed Reznik
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Deaglan McHugh
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Neil J Shah
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Emily Feld
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - David H Aggen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - William Rafelson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Han Xiao
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Maria I Carlo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Darren R Feldman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Chung-Han Lee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Robert J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Martin H Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
175
|
Gervais C, Auclin E, Saltel-Fulero A, Clair G, Oudard S, Mirghani H. Nivolumab immunotherapy rechallenge for progressive laryngeal squamous cell carcinoma after failure of conventional treatment: A CARE case report. Eur Ann Otorhinolaryngol Head Neck Dis 2024; 141:231-234. [PMID: 38418356 DOI: 10.1016/j.anorl.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2024]
Abstract
OBJECTIVE Analysis of rechallenge with nivolumab as 5th-line therapy for locally and nodally failed laryngeal squamous cell carcinoma following conventional therapeutic modalities: radiotherapy, surgery and chemotherapy. OBSERVATION A 70-year-old male, with local and nodal progression of laryngeal squamous cell carcinoma after treatment with chemoradiotherapy and surgery, was initially treated for recurrence with carboplatin, 5-fluorouracile (FU) and cetuximab, followed by second-line nivolumab, and then two lines of conventional chemotherapy with paclitaxel and cetuximab followed by carboplatin and cetuximab. He underwent rechallenge with nivolumab in 5th line, achieving 12months' response, ongoing at the time of writing, and 42.5months' survival since initiation of exclusive systemic management after failure of conventional treatment. CONCLUSION This case report highlights the benefit of nivolumab rechallenge in 5th line following previous failure as stand-alone therapy in 2nd line for a patient with laryngeal squamous cell carcinoma locally and nodally uncontrolled after conventional treatment. Clinical trials evaluating the efficacy of this approach are necessary to assess its contribution, as it is currently not a standard therapeutic option.
Collapse
Affiliation(s)
- C Gervais
- Université Paris Cité, Service d'Oncologie Médicale, Hôpital Européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France.
| | - E Auclin
- Université Paris Cité, Service d'Oncologie Médicale, Hôpital Européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France
| | - A Saltel-Fulero
- Université Paris Cité, Service d'Imagerie, Hôpital Européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France
| | - G Clair
- Université Paris Cité, Service d'Anatomopathologie, Hôpital Européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France
| | - S Oudard
- Université Paris Cité, Service d'Oncologie Médicale, Hôpital Européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France; Inserm U970, PARCC, Paris, France
| | - H Mirghani
- Université Paris Cité, Service d'Oto-Rhino-Laryngologie et Chirurgie Cervicofaciale, Hôpital Européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France
| |
Collapse
|
176
|
Albertini MR, Zuleger CL, Ranheim EA, Shiyanbola O, Sondel PM, Morris ZS, Eickhoff J, Newton MA, Ong IM, Schwartz RW, Hayim R, Kurzman ID, Turek M, Vail DM. Administration of intratumoral GD2-directed interleukin-2 immunocytokine and local radiation therapy to activate immune rejection of spontaneous canine melanoma. Melanoma Res 2024; 34:307-318. [PMID: 38768442 PMCID: PMC11444423 DOI: 10.1097/cmr.0000000000000975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Canine malignant melanoma provides a clinically relevant, large animal parallel patient population to study the GD2-reactive hu14.18-IL-2 immunocytokine as it is similar to human melanoma and expresses GD2. The objectives of this study were to evaluate safety, radiation fractionation, and identify informative biomarkers of an in-situ tumor vaccine involving local radiation therapy plus intratumoral-immunocytokine in melanoma tumor-bearing dogs. Twelve dogs (six dogs/arm) with locally advanced or metastatic melanoma were randomized to receive a single 8 Gy fraction (arm A) or three 8 Gy fractions over 1 week (arm B) to the primary site and regional lymph nodes (when clinically involved) with the single or last fraction 5 days before intratumoral-immunocytokine at 12 mg/m 2 on 3 consecutive days. Serial tumor biopsies were obtained. All 12 dogs completed protocol treatment, and none experienced significant or unexpected adverse events. Evidence of antitumor activity includes one dog with a complete response at day 60, one dog with a partial response at day 60, and four dogs with mixed responses. Histology of serial biopsies shows a variably timed increase in intratumoral lymphocytic inflammation in some dogs. Canine NanoString analyses of serial biopsies identified changes in gene signatures of innate and adaptive cell types versus baseline. There were no significant differences in NanoString results between arm A and arm B. We conclude that intratumoral-immunocytokine in combination with local radiation therapy in canine melanoma is well tolerated and has antitumor activity with the potential to inform clinical development in melanoma patients.
Collapse
Affiliation(s)
- Mark R. Albertini
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Medicine, University of Wisconsin School of Veterinary Medicine
- Department of Dermatology, University of Wisconsin School of Veterinary Medicine
- The Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Cindy L. Zuleger
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Medicine, University of Wisconsin School of Veterinary Medicine
| | - Erik A. Ranheim
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Pathology & Laboratory Medicine, University of Wisconsin School of Veterinary Medicine
| | - Oyewale Shiyanbola
- Stanford University School of Medicine, Department of Pathology, Stanford, California
| | - Paul M. Sondel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Pediatrics, University of Wisconsin School of Veterinary Medicine
- Department of Human Oncology, University of Wisconsin School of Veterinary Medicine
| | - Zachary S. Morris
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Human Oncology, University of Wisconsin School of Veterinary Medicine
| | - Jens Eickhoff
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Biostatistics & Medical Informatics, University of Wisconsin School of Veterinary Medicine
| | - Michael A. Newton
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Biostatistics & Medical Informatics, University of Wisconsin School of Veterinary Medicine
| | - Irene M. Ong
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Biostatistics & Medical Informatics, University of Wisconsin School of Veterinary Medicine
- Department of Obstetrics & Gynecology, University of Wisconsin School of Veterinary Medicine
| | - Rene Welch Schwartz
- Department of Biostatistics & Medical Informatics, University of Wisconsin School of Veterinary Medicine
| | - Rubi Hayim
- Department of Medical Sciences, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Ilene D. Kurzman
- Department of Medical Sciences, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Michelle Turek
- Department of Surgical Sciences, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - David M. Vail
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Medical Sciences, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| |
Collapse
|
177
|
Kanmalar M, Kamal R, Abdul Sani SF, Pathmanathan D, Bm Said NA, Paramanantham Y, Abd Jamil AH, Mun KS, Kuppusamy S, Almugren KS, Almajid HF, Bradley DA. Spectroscopic diagnosis and metabolite characterization of cisplatin resistance regulated by FDFT1 in bladder cancer tissue. Appl Radiat Isot 2024; 210:111372. [PMID: 38810354 DOI: 10.1016/j.apradiso.2024.111372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 11/21/2023] [Accepted: 05/24/2024] [Indexed: 05/31/2024]
Abstract
As is the case for most solid tumours, chemotherapy remains the backbone in the management of metastatic disease. However, the occurrence of chemotherapy resistance is a cause to worry, especially in bladder cancer. Extensive evidence indicates molecular changes in bladder cancer cells to be the underlying cause of chemotherapy resistance, including the reduced expression of farnesyl-diphosphate farnesyltransferase 1 (FDFT1) - a gene involved in cholesterol biosynthesis. This can likely be a hallmark in examining the resistance and sensitivity of chemotherapy drugs. This work performs spectroscopic analysis and metabolite characterization on resistant, sensitive, stable-disease and healthy bladder tissues. Raman spectroscopy has detected peaks at around 1003 cm-1 (squalene), 1178 cm-1 (cholesterol), 1258 cm-1 (cholesteryl ester), 1343 cm-1 (collagen), 1525 cm-1 (carotenoid), 1575 cm-1 (DNA bases) and 1608 cm-1 (cytosine). The peak parameters were examined, and statistical analysis was performed on the peak features, attaining significant differences between the sample groups. Small-angle x-ray scattering (SAXS) measurements observed the triglyceride peak together with 6th, 7th and 8th - order collagen peaks; peak parameters were also determined. Neutron activation analysis (NAA) detected seven trace elements. Carbon (Ca), magnesium (Mg), chlorine (Cl) and sodium (Na) have been found to have the greatest concentration in the sample groups, suggestive of a role as a biomarker for cisplatin resistance studies. Results from the present research are suggested to provide an important insight into understanding the development of drug resistance in bladder cancer, opening up the possibility of novel avenues for treatment through personalised interventions.
Collapse
Affiliation(s)
- M Kanmalar
- Department of Physics, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Raihan Kamal
- Department of Physics, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - S F Abdul Sani
- Department of Physics, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Dharini Pathmanathan
- Department of Mathematic, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Nur Akmarina Bm Said
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | | | - Amira Hajirah Abd Jamil
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - K S Mun
- Department of Pathology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - S Kuppusamy
- Department of Surgery, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - K S Almugren
- Department of Physics, College of Science, Princess Nourah bint Abdulrahman University, Saudi Arabia.
| | - Hadeel F Almajid
- Medicine College, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
| | - D A Bradley
- Sunway University, Centre for Applied Physics and Radiation Technologies, Jalan Universiti, 46150, PJ, Malaysia; School of Mathematics and Physics, University of Surrey, Guildford, GU2 7XH, UK
| |
Collapse
|
178
|
Le UT, Ohm B, Schmid S. [Perioperative Immunotherapy for Resectable Non-Small Cell Lung Cancer: Current Evidence and New Standard of Care]. Zentralbl Chir 2024; 149:S35-S44. [PMID: 39137760 DOI: 10.1055/a-2353-6336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Immunotherapy has drastically changed the treatment of lung cancer not only in systemic disease but also in the perioperative setting in locally advanced non-small cell lung cancer. In particular, the neoadjuvant and perioperative therapy regimes of the CheckMate 816 and KEYNOTE-671 studies as well as the adjuvant therapy according to the IMPower010 and the PEARLS/KEYNOTE-091 protocols have already been approved by the European Medicines Agency (EMA) for the treatment of selected cases. Other therapy protocols and combination therapies with varying drug classes and therapy modalities are currently being examined for their effectiveness and tolerance. The new treatment landscape creates new opportunities but also challenges for the treating disciplines. This article will focus on the current evidence for perioperative immunotherapy for resectable lung cancer and the resulting therapy standards, especially with regard to patient selection for both neoadjuvant and adjuvant immunotherapy, as well as current research efforts.
Collapse
Affiliation(s)
- Uyen-Thao Le
- Klinik für Thoraxchirurgie, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Deutschland
| | - Birte Ohm
- Klinik für Thoraxchirurgie, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Deutschland
| | - Severin Schmid
- Klinik für Thoraxchirurgie, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Deutschland
| |
Collapse
|
179
|
Curigliano G, Jimenez MM, Shimizu T, Keam B, Meric-Bernstam F, Rutten A, Glaspy J, Schuler PJ, Parikh NS, Ising M, Hassounah N, Wu J, Leyk M, Chen X, Burks H, Chaudhury A, Otero J, Cabanas EG. A phase I trial of LHC165 single agent and in combination with spartalizumab in patients with advanced solid malignancies. ESMO Open 2024; 9:103643. [PMID: 39088985 PMCID: PMC11345372 DOI: 10.1016/j.esmoop.2024.103643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 08/03/2024] Open
Abstract
BACKGROUND LHC165 is a Toll-like receptor (TLR)-7 agonist that generates an effective tumor antigen-specific T-cell adaptive immune response as well as durable antitumor responses. We aimed to evaluate the safety, tolerability, efficacy, dose-limiting toxicities, and pharmacokinetics (PK) of LHC165 single agent (SA) ± spartalizumab [PDR001; anti-programmed cell death protein 1 (PD-1)] in adult patients with advanced solid tumors. MATERIALS AND METHODS In this phase I/Ib, open-label, dose-escalation/expansion study, patients received LHC165 SA 100-600 μg biweekly through intratumoral (IT) injection and LHC165 600 μg biweekly + spartalizumab 400 mg Q4W through intravenous (IV) infusion. RESULTS Forty-five patients were enrolled: 21 patients received LHC165 SA, and 24 patients received LHC165 + spartalizumab. The median duration of exposure was 8 weeks (range 2-129 weeks). No maximum tolerated dose was reached. Recommended dose expansion was established as LHC165 600 μg biweekly as SA and in combination with spartalizumab 400 mg Q4W. The most common drug-related adverse events (AEs) were pyrexia (22.2%), pruritus (13.3%), chills (11.1%), and asthenia (4.4%). The only serious AE (SAE) suspected to be related to the study drug was grade 3 pancreatitis (n = 1). Across all tumor types, overall response rate and disease control were 6.7% and 17.8%, respectively. Overall median progression-free survival (PFS) and immune-related PFS was 1.7 months. LHC165 serum PK demonstrated an initial rapid release followed by a slower release due to continued release of LHC165 from the injection site. CONCLUSIONS LHC165 demonstrated acceptable safety and tolerability both as SA and in combination with spartalizumab, and evidence of limited antitumor activity was seen in adult patients with relapsed/refractory or metastatic solid tumors.
Collapse
Affiliation(s)
- G Curigliano
- Istituto Europeo di Oncologia, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| | - M M Jimenez
- Hospital General Universitario Gregorio Maranon, Madrid, Spain
| | - T Shimizu
- National Cancer Center Hospital, Tokyo, Japan
| | - B Keam
- Seoul National University Hospital, Seoul, South Korea
| | - F Meric-Bernstam
- University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - A Rutten
- Sint-Augustinus Hospital, Antwerp, Belgium
| | - J Glaspy
- University of California, Los Angeles, California, USA
| | - P J Schuler
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - N S Parikh
- Biomedical Research, Novartis, East Hanover, New Jersey, USA
| | - M Ising
- Biomedical Research, Novartis, East Hanover, New Jersey, USA
| | - N Hassounah
- Biomedical Research, Novartis, Cambridge, Massachusetts, USA
| | - J Wu
- Biomedical Research, Novartis, Cambridge, Massachusetts, USA
| | - M Leyk
- Biomedical Research, Novartis, Cambridge, Massachusetts, USA
| | - X Chen
- Biomedical Research, Novartis, East Hanover, New Jersey, USA
| | - H Burks
- Biomedical Research, Novartis, Cambridge, Massachusetts, USA
| | - A Chaudhury
- Biomedical Research, Novartis, Cambridge, Massachusetts, USA
| | - J Otero
- Biomedical Research, Novartis, East Hanover, New Jersey, USA
| | - E Garralda Cabanas
- Vall d'Hebron Institute of Oncology (VHIO), Hospital Vall d'Hebron, Barcelona, Spain
| |
Collapse
|
180
|
Fermi M, Botti C, Chiari F, Abeshi A, Presutti L, Miglio M, Mattioli F, Filippini DM, Valerini S, Marchioni D, Molteni G, Serafini E. Squamous cell carcinoma metastatic to the lymph nodes of the parapharyngeal space: case series and systematic review. ACTA OTORHINOLARYNGOLOGICA ITALICA : ORGANO UFFICIALE DELLA SOCIETA ITALIANA DI OTORINOLARINGOLOGIA E CHIRURGIA CERVICO-FACCIALE 2024; 44:223-232. [PMID: 39347547 PMCID: PMC11441514 DOI: 10.14639/0392-100x-n2993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 04/26/2024] [Indexed: 10/01/2024]
Abstract
Objective Parapharyngeal space (PPS) is a rare and unusual site of head and neck squamous cell carcinoma (SCC) metastases. Treatment strategy for PPS metastases is still not well defined. This research aims to investigate the clinical implications and oncological outcomes of SCC metastases in PPS. Material and methods A systematic review was conducted according to PRISMA criteria. The authors considered only articles reporting the history and treatment of patients with PPS SCC metastases. A retrospective chart review was conducted in two tertiary referral academic centers collecting data of patients with diagnosis of PPS SCC metastases between 2010 and 2023 to study their outcome based on clinical presentation and treatment strategy. Results The retrospective chart review showed that the oropharynx was the most frequent primary tumour site. The advanced stage at the time of diagnosis was related to poorer survival and higher recurrence rates. A significant difference in 2-year overall survival in the subgroup of patients who experienced PPS metastases within the primary treatment and those who experienced PPS metastases as regional recurrence (66.7 vs 30.8%) was observed. Similar low survival rates were reported in the literature review with a mean overall and disease-free survival of 19.8 and 8.6 months, respectively. Conclusions PPS metastases are associated with a dismal prognosis, especially when diagnosed as regional recurrence after primary treatment, due to patients' poor general conditions and difficulty of treatment.
Collapse
Affiliation(s)
- Matteo Fermi
- Otorhinolaryngology and Audiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, Università di Bologna, Bologna, Italy
| | - Cecilia Botti
- Otorhinolaryngology and Audiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesco Chiari
- Otorhinolaryngology and Audiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Andi Abeshi
- Otorhinolaryngology and Audiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Livio Presutti
- Otorhinolaryngology and Audiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, Università di Bologna, Bologna, Italy
| | - Matteo Miglio
- Department of Otorhinolaryngology - Head and Neck Surgery, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Francesco Mattioli
- Department of Otorhinolaryngology - Head and Neck Surgery, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Daria Maria Filippini
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, Università di Bologna, Bologna, Italy
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Sara Valerini
- Department of Otorhinolaryngology - Head and Neck Surgery, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Daniele Marchioni
- Department of Otorhinolaryngology - Head and Neck Surgery, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Gabriele Molteni
- Otorhinolaryngology and Audiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, Università di Bologna, Bologna, Italy
| | - Edoardo Serafini
- Department of Otorhinolaryngology - Head and Neck Surgery, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| |
Collapse
|
181
|
Ye W, Hou K, Tao N, Li W, Tan Z, Huang Q, Yang D, Lin H, Deng Z, Xia Y, Yu G. Association between CD4 + T cells ATP levels and disease progression in patients with non‑small cell lung cancer. Oncol Lett 2024; 28:369. [PMID: 38933807 PMCID: PMC11200158 DOI: 10.3892/ol.2024.14502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/19/2024] [Indexed: 06/28/2024] Open
Abstract
Introducing the exploration of stimulated CD4+ cells adenosine triphosphate (sATPCD4) levels for immune monitoring post non-small cell lung cancer (NSCLC) chemotherapy, the present study aimed to investigate its efficacy in gauging the potential risk of disease progression (PD) in patients with NSCLC. Therefore, a total of 89 patients with advanced NSCLC, who underwent chemotherapy between August 15 2022 and August 30 2023 at the Fifth Affiliated Hospital of Guangzhou Medical University (Guangzhou, China), were retrospectively studied. Patients were divided into the PD (n=21) and disease stability (non-PD; n=68) groups and their clinical data were compared. The thresholds for predicting PD were identified using receiver operating characteristics (ROC) curves. Multivariate logistic regression analysis was carried out to assess the association between peripheral blood markers and the incidence of PD. Therefore, post-chemotherapy, significant differences in white blood cell count, non-stimulated CD4+ cells ATP and sATPCD4 levels were obtained between patients in the PD and non-PD groups (P<0.05). In addition, sATPCD4 levels were notably decreased in the PD group compared with the non-PD group. Furthermore, ROC analysis revealed that the predictive threshold for PD was 224.5 ng/ml [area under the curve=0.887; 95% confidence interval, 0.811-0.963]. Additionally, patients with low immunity (ATP <224.5 ng/ml) exhibited a higher risk of PD compared with the high-immunity group (ATP >224.5 ng/ml; P<0.0001). Finally, multivariate logistic regression analysis suggested that sATPCD4 could serve as an independent factor for predicting NSCLC progression. Overall, the current study predicted that immune function could be possibly associated with the risk of PD in patients with NSCLC.
Collapse
Affiliation(s)
- Weipeng Ye
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, Huangpu, Guangzhou 510700, P.R. China
| | - Kailian Hou
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, Huangpu, Guangzhou 510700, P.R. China
| | - Na Tao
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, Huangpu, Guangzhou 510700, P.R. China
| | - Weiyi Li
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, Huangpu, Guangzhou 510700, P.R. China
| | - Zhiqiong Tan
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, Huangpu, Guangzhou 510700, P.R. China
| | - Qunfeng Huang
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, Huangpu, Guangzhou 510700, P.R. China
| | - Dongheng Yang
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, Huangpu, Guangzhou 510700, P.R. China
| | - Haoxin Lin
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, Huangpu, Guangzhou 510700, P.R. China
| | - Zihao Deng
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, Huangpu, Guangzhou 510700, P.R. China
| | - Yuanyuan Xia
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, Huangpu, Guangzhou 510700, P.R. China
| | - Guifang Yu
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, Huangpu, Guangzhou 510700, P.R. China
| |
Collapse
|
182
|
Zhang Z, Yu G, Eresen A, Chen Z, Yu Z, Yaghmai V, Zhang Z. Dendritic cell vaccination combined with irreversible electroporation for treating pancreatic cancer-a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:77. [PMID: 39118942 PMCID: PMC11304422 DOI: 10.21037/atm-23-1882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 02/25/2024] [Indexed: 08/10/2024]
Abstract
Background and Objective Pancreatic ductal adenocarcinoma (PDAC) is 3rd most lethal cancer in the USA leading to a median survival of six months and less than 5% 5-year overall survival (OS). As the only potentially curative treatment, surgical resection is not suitable for up to 90% of the patients with PDAC due to late diagnosis. Highly fibrotic PDAC with an immunosuppressive tumor microenvironment restricts cytotoxic T lymphocyte (CTL) infiltration and functions causing limited success with systemic therapies like dendritic cell (DC)-based immunotherapy. In this study, we investigated the potential benefits of irreversible electroporation (IRE) ablation therapy in combination with DC vaccine therapy against PDAC. Methods We performed a literature search to identify studies focused on DC vaccine therapy and IRE ablation to boost therapeutic response against PDAC indexed in PubMed, Web of Science, and Scopus until February 20th, 2023. Key Content and Findings IRE ablation destructs tumor structure while preserving extracellular matrix and blood vessels facilitating local inflammation. The studies demonstrated IRE ablation reduces tumor fibrosis and promotes CTL tumor infiltration to PDAC tumors in addition to boosting immune response in rodent models. The administration of the DC vaccine following IRE ablation synergistically enhances therapeutic response and extends OS rates compared to the use of DC vaccination or IRE alone. Moreover, the implementation of data-driven approaches further allows dynamic and longitudinal monitoring of therapeutic response and OS following IRE plus DC vaccine immunoablation. Conclusions The combination of IRE ablation and DC vaccine immunotherapy is a potent strategy to enhance the therapeutic outcomes in patients with PDAC.
Collapse
Affiliation(s)
- Zigeng Zhang
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, USA
| | - Guangbo Yu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Aydin Eresen
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, USA
| | - Zhilin Chen
- Department of Human Biology and Business Administration, University of Southern California, Los Angeles, CA, USA
| | - Zeyang Yu
- Information School, University of Washington, Seattle, WA, USA
| | - Vahid Yaghmai
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Zhuoli Zhang
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
183
|
Wang K, Yi H, Lv Z, Jin D, Fu L, Mao Y. Analysis of surgical complexity and short-term prognostic indicators in NSCLC patients: neoadjuvant targeted therapy versus neoadjuvant chemoimmunotherapy. Ther Adv Med Oncol 2024; 16:17588359241265214. [PMID: 39091603 PMCID: PMC11292697 DOI: 10.1177/17588359241265214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/13/2024] [Indexed: 08/04/2024] Open
Abstract
Background Neoadjuvant therapy improves survival benefits in patients with locally advanced non-small cell lung cancer but increases tissue density, presenting challenges for surgeons. Objectives To compare the differences in surgical complexity and short-term prognostic outcomes between neoadjuvant targeted therapy (NTT) and neoadjuvant chemoimmunotherapy (NCI). Design/methods This study enrolled 106 patients underwent curative surgery after neoadjuvant therapy between January 2020 and December 2023 at the National Cancer Center of China. Differences in surgical complexity and short-term prognostic outcomes between the two neoadjuvant therapy cohorts were evaluated. The pathological indicators such as pathological response rate and lymph node upstaging/downstaging were then analyzed. Results In total, 33 patients underwent NTT and 73 underwent NCI preoperatively. Patients who received NTT showed a higher minimally invasive surgery rate (84.8% versus 53.4%, p < 0.01), shorter operative time (144 versus 184 min, p < 0.01), lower conversion rate (3.3% versus 17.8%, p = 0.03), less postoperative drainage (day 3: 140 versus 200 mL, p = 0.03), and lower incidence of postoperative complications including arrhythmias (6.1% versus 26%, p = 0.02). The pathological response rate in the NTT and NCI groups was 70% and 75%, respectively, with the latter group showing a higher complete pathological response rate. The two groups had no significant differences in major pathological response and lymph node pathological response rate. Conclusion Patients who received NTT presented fewer surgical challenges for surgeons and had better surgical outcomes than those who received NCI therapy, with comparable pathological response rates between the two cohorts. Accordingly, NTT is the preferred induction regimen for patients harboring mutation status.
Collapse
Affiliation(s)
- Kun Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hang Yi
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhuoheng Lv
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Donghui Jin
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Fu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yousheng Mao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
184
|
Huang S, Yu H, Li Z, Tang Y, Luo L, Wu J, Li X, Shi Q, Xie S, Qiao G. Electronic patient-reported outcome-based surveillance system to evaluate safety and efficacy of preoperative immunochemotherapy with or without short-term chemoradiation in patients with esophageal squamous cell carcinoma (ePRO-PICCRT): protocol for a prospective, single-arm, phase II study. J Thorac Dis 2024; 16:4719-4726. [PMID: 39144362 PMCID: PMC11320245 DOI: 10.21037/jtd-24-274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/14/2024] [Indexed: 08/16/2024]
Abstract
Background Radiation-associated adverse events (ADEs) in patients with esophageal squamous cell carcinoma (ESCC) remain a problem. Recent research has focused on reducing radiation-associated ADEs while maintaining efficacy, particularly through the combination of immune checkpoint inhibitors (ICIs) with chemotherapy. Patient-reported outcomes (PROs) have also emerged as reliable measures for monitoring treatment effectiveness and quality of life (QoL). This trial aims to investigate the feasibility of using patient-reported dysphagia relief to assess pathological response following neoadjuvant immunochemotherapy, as well as the safety and efficacy of neoadjuvant immunochemotherapy combined with short-course radiotherapy for patients with locally advanced ESCC. Methods This study is designed as a prospective, single-arm, phase II study. Eligible ESCC patients will be invited to participate in this study. All participants will receive paclitaxel (albumin-bound) (260 mg/m2, day 1), carboplatin [area under the curve (AUC) 5; 5 mg/mL/min, day 1] or cisplatin [60 mg/m2, intravenous drip (ivdrip), day 1], and tislelizumab (200 mg, day 1) in the first treatment cycle. Early remission of dysphagia is defined as relief greater than 70% according to the dysphagia symptom score in the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire esophagus-specific questionnaire (EORTC OES-18). The early remission group (Group A) will continue with the same regimen for two treatment cycles. The latent remission group will continue with one treatment cycle followed by neoadjuvant immunochemotherapy combined with short-course radiotherapy (radiotherapy 30 Gy/10 F). The primary objective is the pathological complete response (pCR) rate. Research data collection, storage, and management will be conducted in a web-based Real-World-Data Management Platform (RWDMP). Longitudinal data will be conducted by a linear mixed model with treatment effects, baseline factors influencing the endpoint as fixed effects, and the center as a random effect. Discussion This study will provide evidence for using patient-reported dysphagia relief to evaluate pathological response after neoadjuvant immunochemotherapy in early remission (Group A) and to evaluate the safety and efficacy of combining immunochemotherapy with short-course radiotherapy in latent remission (Group B) among patients with ESCC. Limitations include the single-arm study design, small sample size, and the need for further exploration of the specific mechanism and mediator of early dysphagia remission's effect on immunochemotherapy effectiveness. Trial Registration This study is registered at Clinicaltrials.gov (NCT05596890).
Collapse
Affiliation(s)
- Shujie Huang
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hongfan Yu
- State Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Zijie Li
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yong Tang
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Liling Luo
- Department of Radiation Oncology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Junhan Wu
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xianglin Li
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qiuling Shi
- State Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- State Key Laboratory of Ultrasound Engineering in Medicine, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Songxi Xie
- Department of Radiation Oncology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Guibin Qiao
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
185
|
Eleftheriadou ED, Saroglou M, Syrigos N, Kotteas E, Kouvela M. The role of immunotherapy in patients with lung cancer and brain metastases: a narrative review of the literature. Monaldi Arch Chest Dis 2024. [PMID: 39077863 DOI: 10.4081/monaldi.2024.2967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024] Open
Abstract
Worldwide, approximately half of the patients diagnosed with lung cancer (LC) will develop, simultaneously or asynchronously, brain metastases (BMs). The existence of BMs negatively affects the quality of life and constitutes a poor prognostic factor, linked with high mortality. Locoregional therapy with surgery or radiation is, until now, the treatment of choice, especially for symptomatic patients; however, both options are linked to a high complication rate. The question arising here is whether, in asymptomatic patients, the benefit outweighs the risk and whether an alternative method can be used to treat this special category of patients. Over the last decade, immune checkpoint inhibitors (ICIs) have represented a major breakthrough in the field of oncology, and several molecules have been approved as a treatment option for LC. This review tried to analyze the tumor microenvironment of both the primary lung tumor and the BMs in order to evaluate the intracranial activity of ICIs, outline the main challenges of including these agents in the treatment of LC with BMs, highlight the available information from the main clinical trials, and mark the potential positive effect of choosing a combination therapy. In conclusion, it appears that immunotherapy has a positive effect, inhibiting the progression of BMs, but more data should be published specifically for this category of patients.
Collapse
Affiliation(s)
- Eleni D Eleftheriadou
- Department of Pulmonary Medicine, George Papanikolaou General Hospital, Thessaloniki.
| | - Maria Saroglou
- Department of Pulmonary Medicine, George Papanikolaou General Hospital, Thessaloniki.
| | - Nikolaos Syrigos
- Oncology Unit, 3rd Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens.
| | - Ellias Kotteas
- Oncology Unit, 3rd Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens.
| | - Marousa Kouvela
- Oncology Unit, 3rd Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens.
| |
Collapse
|
186
|
Romero-Ventosa EY, García-Beloso N, López-López A, Martínez-López-de-Castro N. Effectiveness and safety of pembrolizumab as first-line treatment for non-small cell lung cancer in real clinical practice. J Oncol Pharm Pract 2024:10781552241264179. [PMID: 39042931 DOI: 10.1177/10781552241264179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Pembrolizumab is currently the drug for the first-line treatment of stage-IV non-small cell lung cancer. The objective of this study is to measure the effectiveness of pembrolizumab as a first-line treatment and to analyze its safety in real clinical practice. METHODS This was a retrospective study that included patients with metastatic non-small cell lung cancer who had received pembrolizumab as a first-line treatment between 1 June 2018 and 31 January 2021. Variables related to the patients, treatment, and drug's efficacy and safety were collected. RESULTS A total of 50 patients were analyzed. The median real-world progression-free survival and real-world overall survival of those who received pembrolizumab in monotherapy were 10.5 months (95% CI: 2.3-18.6) and 18.9 months (95% CI: 16.9-20.8), respectively. The median real-world progression-free survival and real-world overall survival of those who received the drug with chemotherapy was 7.9 months (95% CI: 4.1-11.7) and 13.3 months (95% CI: 0.0-27), respectively. Mostly digestive (48.3%) and endocrine (41.4%) immune-related adverse events were detected among the patients who received pembrolizumab in monotherapy, whereas mostly digestive immune-related adverse events (85.7%) and hematological toxicities (71.5%) were observed in those treated with pembrolizumab plus chemotherapy. CONCLUSION Pembrolizumab has proven its effectiveness in terms of increasing real-world progression-free survival and real-world overall survival in real clinical practice. The main adverse events were digestive toxicities with pembrolizumab in monotherapy and with chemotherapy.
Collapse
Affiliation(s)
- Elena Yaiza Romero-Ventosa
- Pharmacy Department, University Hospital Complex of Vigo, Vigo, Spain
- Innovation in Clinical Pharmacy Research Group (i-FARMAVigo), Galicia Sur Health Research Institute (IIS Galicia Sur) SERGAS-UVIGO, Vigo, Spain
| | | | - Aida López-López
- Virology and Pathogenesis Group, Galicia Sur Health Research Institute, SERGAS-UVIGO, Vigo, Spain
| | - Noemí Martínez-López-de-Castro
- Pharmacy Department, University Hospital Complex of Vigo, Vigo, Spain
- Innovation in Clinical Pharmacy Research Group (i-FARMAVigo), Galicia Sur Health Research Institute (IIS Galicia Sur) SERGAS-UVIGO, Vigo, Spain
| |
Collapse
|
187
|
Wermke M, Holderried TAW, Luke JJ, Morris VK, Alsdorf WH, Wetzko K, Andersson BS, Wistuba II, Parra ER, Hossain MB, Grund-Gröschke S, Aslan K, Satelli A, Marisetty A, Satam S, Kalra M, Hukelmann J, Kursunel MA, Pozo K, Acs A, Backert L, Baumeister M, Bunk S, Wagner C, Schoor O, Mohamed AS, Mayer-Mokler A, Hilf N, Krishna D, Walter S, Tsimberidou AM, Britten CM. First-in-human dose escalation trial to evaluate the clinical safety and efficacy of an anti-MAGEA1 autologous TCR-transgenic T cell therapy in relapsed and refractory solid tumors. J Immunother Cancer 2024; 12:e008668. [PMID: 39038917 PMCID: PMC11268062 DOI: 10.1136/jitc-2023-008668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2024] [Indexed: 07/24/2024] Open
Abstract
RATIONALE OF THE TRIAL Although the use of engineered T cells in cancer immunotherapy has greatly advanced the treatment of hematological malignancies, reaching meaningful clinical responses in the treatment of solid tumors is still challenging. We investigated the safety and tolerability of IMA202 in a first-in-human, dose escalation basket trial in human leucocyte antigen A*02:01 positive patients with melanoma-associated antigen A1 (MAGEA1)-positive advanced solid tumors. TRIAL DESIGN The 2+2 trial design was an algorithmic design based on a maximally acceptable dose-limiting toxicity (DLT) rate of 25% and the sample size was driven by the algorithmic design with a maximum of 16 patients. IMA202 consists of autologous genetically modified cytotoxic CD8+ T cells expressing a T cell receptor (TCR), which is specific for a nine amino acid peptide derived from MAGEA1. Eligible patients underwent leukapheresis, T cells were isolated, transduced with lentiviral vector carrying MAGEA1-specific TCR and following lymphodepletion (fludarabine/cyclophosphamide), infused with a median of 1.4×109 specific T cells (range, 0.086×109-2.57×109) followed by interleukin 2. SAFETY OF IMA202: No DLT was observed. The most common grade 3-4 adverse events were cytopenias, that is, neutropenia (81.3%), lymphopenia (75.0%), anemia (50.0%), thrombocytopenia (50.0%) and leukopenia (25.0%). 13 patients experienced cytokine release syndrome, including one grade 3 event. Immune effector cell-associated neurotoxicity syndrome was observed in two patients and was grade 1 in both. EFFICACY OF IMA202: Of the 16 patients dosed, 11 (68.8%) patients had stable disease (SD) as their best overall response (Response Evaluation Criteria in Solid Tumors V.1.1). Five patients had initial tumor shrinkage in target lesions and one patient with SD experienced continued shrinkage in target lesions for 3 months in total but had to be classified as progressive disease due to progressive non-target lesions. IMA202 T cells were persistent in peripheral blood for several weeks to months and were also detectable in tumor tissue. Peak persistence was higher in patients who received higher doses. CONCLUSION In conclusion, IMA202 had a manageable safety profile, and it was associated with biological and potential clinical activity of MAGEA1-targeting genetically engineered TCR-T cells in a poor prognosis, multi-indication solid tumor cohort. TRIAL REGISTRATION NUMBERS NCT04639245, NCT05430555.
Collapse
Affiliation(s)
- Martin Wermke
- Department of Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
- National Center for Tumor Diseases, Dresden, Germany
| | - Tobias A W Holderried
- Department of Hematology, Oncology, Immunooncology, Stem Cell Transplantation, and Rheumatology, University Hospital Bonn, Bonn, Germany
| | - Jason John Luke
- Cancer Immunotherapeutics Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Van K Morris
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center Division of Cancer Medicine, Houston, Texas, USA
| | - Winfried H Alsdorf
- Department of Oncology, Hematology, and Bone Marrow Transplantation with Section Pneumology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Katrin Wetzko
- Department of Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Borje S Andersson
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Edwin R Parra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | - Katrin Aslan
- Immatics Biotechnologies GmbH, Tuebingen, Germany
| | | | | | - Swapna Satam
- Immatics Biotechnologies GmbH, Tuebingen, Germany
| | | | | | | | | | - Andreas Acs
- Immatics Biotechnologies GmbH, Tuebingen, Germany
| | | | | | | | | | | | | | | | - Norbert Hilf
- Immatics Biotechnologies GmbH, Tuebingen, Germany
| | | | | | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | |
Collapse
|
188
|
Glazer SE, Kummar S, Mittra E. Illuminating immunotherapy response via precision T cell-targeted PET imaging. Front Med (Lausanne) 2024; 11:1233913. [PMID: 39104861 PMCID: PMC11298440 DOI: 10.3389/fmed.2024.1233913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 06/20/2024] [Indexed: 08/07/2024] Open
Abstract
Traditionally, immunotherapy agent selection and treatment strategies are guided by biopsy-based histological information. However, biopsies are limited in that they are invasive, provide static information regarding the tumor immune microenvironment, and only sample a small part of one tumor site. The tumor microenvironment is dynamic and heterogenous. As a result, the immune milieu at one site may be distinct from other metastatic sites. These factors make identifying which patients are likely to respond to different immunotherapies and which harbor intrinsic resistance mechanisms difficult to identify based on a biopsy alone. As such, there is significant interest in alternative methodologies that better characterize the tumor immune microenvironment and monitor immunotherapy response. PET imaging potentially offers a non-invasive way to characterize the tumor immune microenvironment at the primary tumor and metastases and allow for longitudinal characterization. Herein, we review pre-clinically and clinically tested T cell-targeted PET radiopharmaceuticals, as T cells have been the dominant immunotherapy target, and their utility in both evaluating response to immunotherapy and in understanding the systemic immune response to treatment with immunotherapeutics.
Collapse
Affiliation(s)
- Sarah E. Glazer
- Division of Internal Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Shivaani Kummar
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Erik Mittra
- Division of Molecular Imaging and Therapy, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
189
|
Mulier G, Lin R, Aparicio T, Biard L. Bayesian sequential monitoring strategies for trials of digestive cancer therapeutics. BMC Med Res Methodol 2024; 24:154. [PMID: 39030498 PMCID: PMC11526600 DOI: 10.1186/s12874-024-02278-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND New therapeutics in oncology have presented challenges to existing paradigms and trial designs in all phases of drug development. As a motivating example, we considered an ongoing phase II trial planned to evaluate the combination of a MET inhibitor and an anti-PD-L1 immunotherapy to treat advanced oesogastric carcinoma. The objective of the paper was to exemplify the planning of an adaptive phase II trial with novel anti-cancer agents, including prolonged observation windows and joint sequential evaluation of efficacy and toxicity. METHODS We considered various candidate designs and computed decision rules assuming correlations between efficacy and toxicity. Simulations were conducted to evaluate the operating characteristics of all designs. RESULTS Design approaches allowing continuous accrual, such as the time-to-event Bayesian Optimal Phase II design (TOP), showed good operating characteristics while ensuring a reduced trial duration. All designs were sensitive to the specification of the correlation between efficacy and toxicity during planning, but TOP can take that correlation into account more easily. CONCLUSIONS While specifying design working hypotheses requires caution, Bayesian approaches such as the TOP design had desirable operating characteristics and allowed incorporating concomittant information, such as toxicity data from concomitant observations in another relevant patient population (e.g., defined by mutational status).
Collapse
Affiliation(s)
- Guillaume Mulier
- ECSTRRA team UMR 1153, INSERM, Saint-Louis hospital, 1 avenue Claude Vellefaux, Paris, 75010, France.
- Service de Biostatistique et Information Médicale, AP-HP Saint-Louis hospital, 1 avenue Claude Vellefaux, Paris, 75010, France.
| | - Ruitao Lin
- Department of Biostatistics, MD Anderson Cancer Center, 7007 Bertner Avenue, Houston, 77030, Texas, USA
| | - Thomas Aparicio
- Service d'hépato-gastro-entérologie, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, Paris, 75010, France
- Université Paris Cité, 12 rue de l'École-de-Médecine, Paris, 75006, France
| | - Lucie Biard
- ECSTRRA team UMR 1153, INSERM, Saint-Louis hospital, 1 avenue Claude Vellefaux, Paris, 75010, France
- Service de Biostatistique et Information Médicale, AP-HP Saint-Louis hospital, 1 avenue Claude Vellefaux, Paris, 75010, France
| |
Collapse
|
190
|
Dvir K, Giordano S, Leone JP. Immunotherapy in Breast Cancer. Int J Mol Sci 2024; 25:7517. [PMID: 39062758 PMCID: PMC11276856 DOI: 10.3390/ijms25147517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Breast cancer is a disease encompassing a spectrum of molecular subtypes and clinical presentations, each with distinct prognostic implications and treatment responses. Breast cancer has traditionally been considered an immunologically "cold" tumor, unresponsive to immunotherapy. However, clinical trials in recent years have found immunotherapy to be an efficacious therapeutic option for select patients. Breast cancer is categorized into different subtypes ranging from the most common positive hormone receptor (HR+), human epidermal growth factor receptor 2 (HER2)-negative type, to less frequent HER2- positive breast cancer and triple-negative breast cancer (TNBC), highlighting the necessity for tailored treatment strategies aimed at maximizing patient outcomes. Despite notable progress in early detection and new therapeutic modalities, breast cancer remains the second leading cause of cancer death in the USA. Moreover, in recent decades, breast cancer incidence rates have been increasing, especially in women younger than the age of 50. This has prompted the exploration of new therapeutic approaches to address this trend, offering new therapeutic prospects for breast cancer patients. Immunotherapy is a class of therapeutic agents that has revolutionized the treatment landscape of many cancers, namely melanoma, lung cancer, and gastroesophageal cancers, amongst others. Though belatedly, immunotherapy has entered the treatment armamentarium of breast cancer, with the approval of pembrolizumab in combination with chemotherapy in triple-negative breast cancer (TNBC) in the neoadjuvant and advanced settings, thereby paving the path for further research and integration of immune checkpoint inhibitors in other subtypes of breast cancer. Trials exploring various combination therapies to harness the power of immunotherapy in symbiosis with various chemotherapeutic agents are ongoing in hopes of improving response rates and prolonging survival for breast cancer patients. Biomarkers and precise patient selection for the utilization of immunotherapy remain cardinal and are currently under investigation, with some biomarkers showing promise, such as Program Death Lignat-1 (PDL-1) Combined Positive Score, Tumor Mutation Burden (TMB), and Tumor Infiltrating Lymphocytes (TILs). This review will present the current landscape of immunotherapy, particularly checkpoint inhibitors, in different types of breast cancer.
Collapse
Affiliation(s)
- Kathrin Dvir
- Dana Farber Cancer Institute, Boston, MA 02215, USA; (K.D.)
- St. Elizabeth’s Medical Center, Boston, MA 02111, USA
| | - Sara Giordano
- Dana Farber Cancer Institute, Boston, MA 02215, USA; (K.D.)
- St. Elizabeth’s Medical Center, Boston, MA 02111, USA
| | | |
Collapse
|
191
|
Chen Y, Wang D, Li Y, Qi L, Si W, Bo Y, Chen X, Ye Z, Fan H, Liu B, Liu C, Zhang L, Zhang X, Li Z, Zhu L, Wu A, Zhang Z. Spatiotemporal single-cell analysis decodes cellular dynamics underlying different responses to immunotherapy in colorectal cancer. Cancer Cell 2024; 42:1268-1285.e7. [PMID: 38981439 DOI: 10.1016/j.ccell.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 04/10/2024] [Accepted: 06/14/2024] [Indexed: 07/11/2024]
Abstract
Expanding the efficacy of immune checkpoint blockade (ICB) in colorectal cancer (CRC) presses for a comprehensive understanding of treatment responsiveness. Here, we analyze multiple sequential single-cell samples from 22 patients undergoing PD-1 blockade to map the evolution of local and systemic immunity of CRC patients. In tumors, we identify coordinated cellular programs exhibiting distinct response associations. Specifically, exhausted T (Tex) or tumor-reactive-like CD8+ T (Ttr-like) cells are closely related to treatment efficacy, and Tex cells show correlated proportion changes with multiple other tumor-enriched cell types following PD-1 blockade. In addition, we reveal the less-exhausted phenotype of blood-associated Ttr-like cells in tumors and find that their higher abundance suggests better treatment outcomes. Finally, a higher major histocompatibility complex (MHC) II-related signature in circulating CD8+ T cells at baseline is linked to superior responses. Our study provides insights into the spatiotemporal cellular dynamics following neoadjuvant PD-1 blockade in CRC.
Collapse
Affiliation(s)
- Yuqing Chen
- Biomedical Pioneering Innovative Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Dongfang Wang
- Biomedical Pioneering Innovative Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China.
| | - Yingjie Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Unit III, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lu Qi
- Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing, China
| | - Wen Si
- Biomedical Pioneering Innovative Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Yufei Bo
- Biomedical Pioneering Innovative Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Xueyan Chen
- Biomedical Pioneering Innovative Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Zhaochen Ye
- Biomedical Pioneering Innovative Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Hongtao Fan
- Biomedical Pioneering Innovative Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Baolin Liu
- Biomedical Pioneering Innovative Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Chang Liu
- Biomedical Pioneering Innovative Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Li Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Pathology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiaoyan Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Radiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhongwu Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Pathology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Linna Zhu
- Biomedical Pioneering Innovative Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Aiwen Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Unit III, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovative Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
192
|
Takenaka Y, Takemoto N, Otsuka T, Nishio M, Tanida M, Fujii T, Hayashi K, Suzuki M, Mori M, Yamamoto Y, Uno A, Inohara H. Validation and comparison of prognostic scoring systems in patients with head and neck squamous cell carcinoma treated with nivolumab. Jpn J Clin Oncol 2024; 54:761-769. [PMID: 38555496 DOI: 10.1093/jjco/hyae042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 03/13/2024] [Indexed: 04/02/2024] Open
Abstract
OBJECTIVE Several scoring systems have been developed to predict prognosis in patients with refractory cancer. We aimed to validate eight scoring systems and determine the best method for predicting the prognosis of head and neck squamous cell carcinoma treated with nivolumab. METHODS This multicentre retrospective study involved 154 patients with recurrent and/or metastatic head and neck squamous cell carcinoma treated with nivolumab between 2017 and 2020. Oncological outcomes were assessed according to the scoring systems, including MD Anderson Cancer Center + neutrophil-to-lymphocyte ratio and Hammersmith scores. Objective response, overall survival and progression-free survival were evaluated using logistic regression and Cox proportional hazards analyses. Receiver operating curve analysis was used to calculate the area under the curve and estimate the efficacy of each score. RESULTS No significant associations were found between the responses and any score. Seven of the eight scoring systems were associated with disease control (odds ratio, 0.26-0.70). Amongst the eight scoring systems, MD Anderson Cancer Center + neutrophil-to-lymphocyte ratio showed the highest area under the curve for predicting response and disease control. Seven scoring systems were prognostic factors for progression-free survival (hazard ratio, 1.22-1.95). All eight scoring systems were prognostic factors for overall survival (hazard ratio, 1.62-3.83). According to the time-dependent receiver operating characteristics analysis for overall survival, the Hammersmith scoring system had the best predictive ability at 3 months, and the MD Anderson Cancer Center + neutrophil-to-lymphocyte ratio scoring system had the highest area under the curve between 6 and 24 months. CONCLUSIONS MD Anderson Cancer Center + neutrophil-to-lymphocyte ratio and Hammersmith scoring systems were better predictors of prognosis in patients with head and neck squamous cell carcinoma treated with nivolumab.
Collapse
Affiliation(s)
- Yukinori Takenaka
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Norihiko Takemoto
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomoyuki Otsuka
- Department of Medical Oncology, International Cancer Institute, Osaka, Japan
| | - Minako Nishio
- Department of Medical Oncology, International Cancer Institute, Osaka, Japan
| | - Masashi Tanida
- Department of Head and Neck Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Takashi Fujii
- Department of Head and Neck Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Kazuki Hayashi
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Motoyuki Suzuki
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masashi Mori
- Department of Otorhinolaryngology-Head and Neck Surgery, General Medical Center, Osaka, Japan
| | - Yoshifumi Yamamoto
- Department of Otorhinolaryngology-Head and Neck Surgery, General Medical Center, Osaka, Japan
| | - Atsuhiko Uno
- Department of Otorhinolaryngology-Head and Neck Surgery, General Medical Center, Osaka, Japan
| | - Hidenori Inohara
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
193
|
Marchal C, Orillard E, Calais F, Westeel V. Immunotherapy for non-small cell lung cancer in the elderly population: a generic protocol. Cochrane Database Syst Rev 2024; 7:CD014907. [PMID: 38958139 PMCID: PMC11220895 DOI: 10.1002/14651858.cd014907.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
OBJECTIVES This is a protocol for a Cochrane Review (intervention). The objectives are as follows: To evaluate the effectiveness and safety of immune checkpoint inhibitors (ICI) as monotherapy or in combination compared to standard of care for elderly people (≥ 65 years) with non-small cell lung cancer (NSCLC).
Collapse
Affiliation(s)
| | - Emeline Orillard
- Université Bourgogne Franche-Comté, INSERM, EFS BFC, UMR 1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - François Calais
- Bibliothèque universitaire de Santé, Université de Franche-Comté, Besançon, France
| | - Virginie Westeel
- Université Bourgogne Franche-Comté, INSERM, EFS BFC, UMR 1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Department of Thoracic Oncology, University Hospital of Besançon, Besançon, France
| |
Collapse
|
194
|
Yu JH, Xiao BY, Li DD, Jiang W, Ding Y, Wu XJ, Zhang RX, Lin JZ, Wang W, Han K, Kong LH, Zhang XK, Chen BY, Mei WJ, Pan ZZ, Tang JH, Zhang XS, Ding PR. Neoadjuvant camrelizumab plus apatinib for locally advanced microsatellite instability-high or mismatch repair-deficient colorectal cancer (NEOCAP): a single-arm, open-label, phase 2 study. Lancet Oncol 2024; 25:843-852. [PMID: 38852601 DOI: 10.1016/s1470-2045(24)00203-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND PD-1 blockade is highly efficacious for mismatch repair-deficient colorectal cancer in both metastatic and neoadjuvant settings. We aimed to explore the activity and safety of neoadjuvant therapy with PD-1 blockade plus an angiogenesis inhibitor and the feasibility of organ preservation in patients with locally advanced mismatch repair-deficient colorectal cancer. METHODS We initiated a single-arm, open-label, phase 2 trial (NEOCAP) at Sun Yat-sen University Cancer Center and the Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China. Patients aged 18-75 years with untreated mismatch repair-deficient or microsatellite instability-high or POLE/POLD1-mutated locally advanced colorectal cancer (cT3 or N+ for rectal cancer, and T3 with invasion ≥5mm or T4, with or without N+ for colon cancer) and an Eastern Cooperative Oncology Group performance score of 0-1 were enrolled and given 200 mg camrelizumab intravenously on day 1 and 250 mg apatinib orally from day 1-14, every 3 weeks for 3 months followed by surgery or 6 months if patients did not have surgery. Patients who had a clinical complete response did not undergo surgery and proceeded with a watch-and-wait approach. The primary endpoint was the proportion of patients with a pathological or clinical complete response. Eligible enrolled patients who received at least one cycle of neoadjuvant treatment and had at least one tumour response assessment following the baseline assessment were included in the activity analysis, and patients who received at least one dose of study drug were included in the safety analysis. The study is registered with ClinicalTrials.gov (NCT04715633) and is ongoing. FINDINGS Between Sept 29, 2020, and Dec 15, 2022, 53 patients were enrolled; one patient was excluded from the activity analysis because they were found to be mismatch repair-proficient and microsatellite-stable. 23 (44%) patients were female and 29 (56%) were male. The median follow-up was 16·4 (IQR 10·5-23·5) months. 28 (54%; 95% CI 35-68) patients had a clinical complete response and 24 of these patients were managed with a watch-and-wait approach, including 20 patients with colon cancer and multiple primary colorectal cancer. 23 (44%) of 52 patients underwent surgery for the primary tumour, and 14 (61%; 95% CI 39-80) had a pathological complete response. 38 (73%; 95% CI 59-84) of 52 patients had a complete response. Grade 3-5 adverse events occurred in 20 (38%) of 53 patients; the most common were increased aminotransferase (six [11%]), bowel obstruction (four [8%]), and hypertension (four [8%]). Drug-related serious adverse events occurred in six (11%) of 53 patients. One patient died from treatment-related immune-related hepatitis. INTERPRETATION Neoadjuvant camrelizumab plus apatinib show promising antitumour activity in patients with locally advanced mismatch repair-deficient or microsatellite instability-high colorectal cancer. Immune-related adverse events should be monitored with the utmost vigilance. Organ preservation seems promising not only in patients with rectal cancer, but also in those with colon cancer who have a clinical complete response. Longer follow-up is needed to assess the oncological outcomes of the watch-and-wait approach. FUNDING The National Natural Science Foundation of China, Guangdong Basic and Applied Basic Research Foundation, and the Cancer Innovative Research Program of Sun Yat-sen University Cancer Center. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Jie-Hai Yu
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Bin-Yi Xiao
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Dan-Dan Li
- Department of Biotherapy Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Wu Jiang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Ya Ding
- Department of Biotherapy Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Xiao-Jun Wu
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Rong-Xin Zhang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Jun-Zhong Lin
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Wei Wang
- Department of Gastrointestinal Surgery, Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China; Department of General Surgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Kai Han
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Ling-Heng Kong
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Xin-Ke Zhang
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Bi-Yun Chen
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Wei-Jian Mei
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Zhi-Zhong Pan
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Jing-Hua Tang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Xiao-Shi Zhang
- Department of Biotherapy Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Pei-Rong Ding
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China.
| |
Collapse
|
195
|
Jiménez-Labaig P, Rullan A, Hernando-Calvo A, Llop S, Bhide S, O'Leary B, Braña I, Harrington KJ. A systematic review of antibody-drug conjugates and bispecific antibodies in head and neck squamous cell carcinoma and nasopharyngeal carcinoma: Charting the course of future therapies. Cancer Treat Rev 2024; 128:102772. [PMID: 38820656 DOI: 10.1016/j.ctrv.2024.102772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/02/2024]
Abstract
INTRODUCTION There is a need to improve the outcomes of patients with head and neck squamous cell carcinoma (HNSCC) and nasopharyngeal carcinoma (NPC), especially in recurrent unresectable and metastatic (R/M) setting. Antibody-drug conjugates (ADC) and bispecific antibodies (BsAb) may deliver promising results. METHODS We conducted a systematic literature review to identify ADC and BsAb clinical trials, involving patients with HNSCC and NPC, from database creation to December 2023. We reported trial characteristics, overall response rate (ORR), overall survival (OS), and grade ≥ 3 treatment-related adverse events (trAEs). RESULTS 23 trials (65 % phase I) were found, involving 540 R/M patients (355 [20trials] HNSCC and 185 [5trials] NPC). There were 13 ADC (n = 343) and 10 BsAb (n = 197) trials. 96 % patients were refractory to standard of care treatments. ORR ranged from 0 to 100 %, with the highest ORR for GEN1042 plus chemoimmunotherapy. ORRs for monotherapies were 47 % for ADC, and 0-37 % for BsAb. MRG003 reached in HNSCC 43 % and NPC 47 %. BL-B01D1 54 % in NPC. Longest median OS was seen with MRG003 and KN046. Grade ≥ 3 trAEs were 28-60 % in ADC trials, and 3-33 % BsAb. Grade ≥ 3 myelosuppressive trAEs were typically seen in 8 ADC trials, while 4 BsAb showed infusion-related reactions (IRR). Four treatment-related deaths were reported (1 pneumonitis), all ADC trials. CONCLUSION ADC and BsAb antibodies show promise in R/M HNSCC and NPC. Results are premature by small sample sizes and lack of control arm. ADC mainly caused myelosuppression and a pneumonitis case, and BsAb IRR. Further research is warranted in this setting.
Collapse
Affiliation(s)
- Pablo Jiménez-Labaig
- Head and Neck Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom; The Institute of Cancer Research, Division of Radiotherapy and Imaging, London, United Kingdom
| | - Antonio Rullan
- Head and Neck Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom; The Institute of Cancer Research, Division of Radiotherapy and Imaging, London, United Kingdom
| | - Alberto Hernando-Calvo
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Early Phase Clinical Trials Unit (UITM), Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Sandra Llop
- Head and Neck Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Shreerang Bhide
- Head and Neck Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom; The Institute of Cancer Research, Division of Radiotherapy and Imaging, London, United Kingdom
| | - Ben O'Leary
- Head and Neck Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom; The Institute of Cancer Research, Division of Radiotherapy and Imaging, London, United Kingdom
| | - Irene Braña
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Early Phase Clinical Trials Unit (UITM), Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; Lung and Head & Neck Tumors Unit, Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Kevin J Harrington
- Head and Neck Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom; The Institute of Cancer Research, Division of Radiotherapy and Imaging, London, United Kingdom
| |
Collapse
|
196
|
Lim AM, Le Tourneau C, Hurt C, Laskar SG, Steuer CE, Chow VLY, Szturz P, Henson C, Day AT, Bates JE, Lazarakis S, McDowell L, Mehanna H, Yom SS. Assessment of endpoint definitions in recurrent and metastatic mucosal head and neck squamous cell carcinoma trials: Head and Neck Cancer International Group consensus recommendations. Lancet Oncol 2024; 25:e308-e317. [PMID: 38936389 DOI: 10.1016/s1470-2045(24)00068-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 06/29/2024]
Abstract
Transparent and precise endpoint definitions are a crucial aspect of clinical trial conduct and reporting, and are used to communicate the benefit of an intervention. Previous studies have identified inconsistencies in endpoint definitions across oncological clinical trials. Here, the Head and Neck Cancer International Group assessed endpoint definitions from phase 3 trials or trials considered practice-changing for patients with recurrent or metastatic mucosal head and neck squamous cell carcinoma, published between 2008 and 2021. We identify considerable and global heterogeneity in endpoint definitions, which undermines the interpretation of results and development of future studies. We show how fundamental components of even incontrovertible endpoints such as overall survival vary widely, highlighting an urgent need for increased rigour in reporting and harmonisation of endpoints.
Collapse
Affiliation(s)
- Annette M Lim
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia.
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France; INSERM U900 Research unit, Institut Curie, Paris, France; Paris-Saclay University, Paris, France
| | - Chris Hurt
- Southampton Clinical Trials Unit, University of Southampton, Southampton, UK
| | - Sarbani G Laskar
- Department of Radiation Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Conor E Steuer
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Velda L Y Chow
- Division of Head and Neck Surgery, Department of Surgery, The University of Hong Kong, Queen Mary Hospital, Pok Fu Lam, Hong Kong Special Administrative Region, China
| | - Petr Szturz
- Department of Oncology, University of Lausanne and Lausanne University Hospital, Lausanne, Switzerland
| | - Christina Henson
- Department of Radiation Oncology, Stephenson Cancer Center, University of Oklahoma, OK, USA
| | - Andrew T Day
- Department of Otolaryngology-Head and Neck Surgery, University of Texas Southwestern Medical Center, TX, USA
| | - James E Bates
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Smaro Lazarakis
- Health Sciences Library, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Lachlan McDowell
- Department of Radiation Oncology, Princess Alexandra Hospital, Brisbane, QLD, Australia; Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Hisham Mehanna
- Institute of Head and Neck Studies and Education (InHANSE), University of Birmingham, Edgbaston, Birmingham, UK
| | - Sue S Yom
- Department of Radiation Oncology, University of California, San Francisco, CA, USA; Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, CA, USA.
| |
Collapse
|
197
|
Sheth RA, Wehrenberg-Klee E, Patel SP, Brock KK, Fotiadis N, de Baère T. Intratumoral Injection of Immunotherapeutics: State of the Art and Future Directions. Radiology 2024; 312:e232654. [PMID: 39078294 PMCID: PMC11294769 DOI: 10.1148/radiol.232654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 02/17/2024] [Accepted: 02/28/2024] [Indexed: 07/31/2024]
Abstract
Systemic immunotherapies have led to tremendous progress across the cancer landscape. However, several challenges exist, potentially limiting their efficacy in the treatment of solid tumors. Direct intratumoral injection can increase the therapeutic index of immunotherapies while overcoming many of the barriers associated with systemic administration, including limited bioavailability to tumors and potential systemic safety concerns. However, challenges remain, including the lack of standardized approaches for administration, issues relating to effective drug delivery, logistical hurdles, and safety concerns specific to this mode of administration. This article reviews the biologic rationale for the localized injection of immunotherapeutic agents into tumors. It also addresses the existing limitations and practical considerations for safe and effective implementation and provide recommendations for optimizing logistics and treatment workflows. It also highlights the critical role that radiologists, interventional radiologists, and medical physicists play in intratumoral immunotherapy with respect to target selection, image-guided administration, and response assessment.
Collapse
Affiliation(s)
- Rahul A. Sheth
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Eric Wehrenberg-Klee
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Sapna P. Patel
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Kristy K. Brock
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Nicos Fotiadis
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Thierry de Baère
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| |
Collapse
|
198
|
Salari K, Lee JS, Ye H, Seymour ZA, Lee KC, Chinnaiyan P, Grills IS. Long-term survival in patients with brain-only metastatic non-small cell lung cancer undergoing upfront intracranial stereotactic radiosurgery and definitive treatment to the thoracic primary site. Radiother Oncol 2024; 196:110262. [PMID: 38556172 DOI: 10.1016/j.radonc.2024.110262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/21/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND AND PURPOSE To evaluate modern clinical outcomes for patients with brain-only metastatic non-small cell lung cancer (NSCLC) treated with intracranial stereotactic radiosurgery (SRS) with or without definitive treatment of the primary site. MATERIALS AND METHODS Patients with synchronously diagnosed NSCLC and brain-only metastatic disease treated with intracranial SRS at a single institution were retrospectively identified. Patients were stratified based on whether they did (A) or did not (B) receive definitive primary site treatment. Patient characteristics and clinical outcomes were compared. RESULTS From 2008 to 2022, 103 patients were identified, 53 of whom received definitive primary site treatment. Median follow-up was 2.1 y (A) and 0.8 y (B) (p < 0.001). 28 (53 %) patients in Group A received immune checkpoint inhibitor (ICI) therapy versus 19 (38 %) in Group B (p = 0.13) and there were no other statistically significant baseline or treatment characteristic differences between the groups. 5-year local-PFS was 34.5 % (A) versus 0 % (B) (p < 0.001). 5-year regional-PFS was 33.0 % (A) versus 0 % (B) (p < 0.001). 5-year distant body-PFS was 34.0 % (A) versus 0 % (B) (p < 0.001). 5-year CNS-PFS was 14.7 % (A) versus 0 % (B) (p = 0.12). 5-year OS was 40.2 % (A) versus 0 % (B) (p = 0.001). 5-year CSS was 67.6 % (A) versus 0 % (B) (p = 0.002). On multivariable analysis, lack of definitive treatment to the primary site (HR = 2.40), AJCC T3-4 disease (HR = 2.73), and lack of ICI therapy (HR = 2.86) were significant predictors of death. CONCLUSION Definitive treatment to the thoracic primary site in patients with brain-only metastatic NSCLC after intracranial radiosurgery was associated with slower progression of disease and improved survival.
Collapse
Affiliation(s)
- K Salari
- Department of Radiation Oncology, Corewell Health East William Beaumont University Hospital, Royal Oak, MI, United States.
| | - J S Lee
- Department of Radiation Oncology, Corewell Health East William Beaumont University Hospital, Royal Oak, MI, United States
| | - H Ye
- Department of Radiation Oncology, Corewell Health East William Beaumont University Hospital, Royal Oak, MI, United States
| | - Z A Seymour
- Department of Radiation Oncology, Corewell Health Dearborn Hospital, Dearborn, MI, United States
| | - K C Lee
- Department of Radiation Oncology, Corewell Health Troy Hospital, Troy, MI, United States
| | - P Chinnaiyan
- Department of Radiation Oncology, Corewell Health East William Beaumont University Hospital, Royal Oak, MI, United States
| | - I S Grills
- Department of Radiation Oncology, Corewell Health East William Beaumont University Hospital, Royal Oak, MI, United States
| |
Collapse
|
199
|
Uchimoto T, Matsuda T, Komura K, Fukuokaya W, Adachi T, Hirasawa Y, Hashimoto T, Yoshizawa A, Saruta M, Hashimoto M, Higashio T, Tsuchida S, Nishimura K, Tsujino T, Nakamura K, Fukushima T, Nishio K, Yamamoto S, Iwatani K, Urabe F, Mori K, Yanagisawa T, Tsuduki S, Takahara K, Inamoto T, Miki J, Fujita K, Kimura T, Ohno Y, Shiroki R, Uemura H, Azuma H. C-Reactive Protein-Albumin Ratio Predicts Objective Response to Enfortumab Vedotin in Metastatic Urothelial Carcinoma. Target Oncol 2024; 19:635-644. [PMID: 38807017 DOI: 10.1007/s11523-024-01068-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Enfortumab vedotin (EV), an antibody-drug conjugate that targets Nectin-4, is used for patients with metastatic urothelial carcinoma who have experienced progression on platinum-based chemotherapy and checkpoint inhibitors. Despite the widespread use of the drug, evidence remains scarce regarding clinical indicators that can predict the response to EV treatment. OBJECTIVE We aimed to explore the predictive value of clinical indicators derived from peripheral blood tests for treatment responses to EV. METHODS We utilized records of 109 patients with metastatic urothelial carcinoma treated by EV from our multi-institutional dataset. Receiver operating characteristic curve analyses for predicting objective responses including several indicators from blood examinations, such as C-reactive protein-albumin ratio (CAR), hemoglobin, neutrophil-lymphocyte ratio, platelet-lymphocyte ratio, and lactate dehydrogenase, were performed. The optimal cutoff points were determined by the Youden index. Logistic regression analyses for achieving objective responses to EV treatment were performed among these indicators. RESULTS The median age of the cohort was 74 years, and the median follow-up duration was 10 months for the entire group. Median overall survival and progression-free survival from the initiation of EV were 12 and 6 months, respectively. The objective response rate and disease control rate were 48% and 70%, respectively. The receiver operating characteristic curve analysis aimed at predicting the achievement of an objective response to EV showed that the concordant index for the CAR was 0.774, significantly surpassing other indicators such as hemoglobin level, neutrophil-lymphocyte ratio, platelet-lymphocyte ratio, and serum lactate dehydrogenase. The Youden index identified an optimal cutoff value of 1 for CAR (mg/L for C-reactive protein and g/dL for serum albumin level) in predicting the objective response to EV treatment. Using the cutoff value for the CAR, the cohort was divided into 32 patients (29%) with lower CAR and 77 patients (71%) with higher CAR. The objective response rate was observed to be 84% in the lower CAR group and 32% in the higher CAR group (p < 0.0001). A logistic regression analysis revealed that an Eastern Cooperative Oncology Group Performance Status ≥1 (p = 0.04) and a CAR ≥1 (p < 0.001) were identified as independent predictors for the objective response to EV. CONCLUSIONS The evaluation of the CAR from a concise blood examination at the initiation of EV could effectively predict the treatment response to EV in patients with metastatic urothelial carcinoma after the progression of platinum-based chemotherapy and checkpoint inhibitors.
Collapse
Affiliation(s)
- Taizo Uchimoto
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Takuya Matsuda
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Kazumasa Komura
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan.
- Division of Translational Research, Department of Urology, Osaka Medical and Pharmaceutical University, Daigaku-machi 2-7, Takatsuki, Japan.
| | - Wataru Fukuokaya
- Department of Urology, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Takahiro Adachi
- Department of Urology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Yosuke Hirasawa
- Department of Urology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Takeshi Hashimoto
- Department of Urology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Atsuhiko Yoshizawa
- Department of Urology, Fujita-Health University School of Medicine, Kutsukake, Toyoake, Aichi Nagoya, Japan
| | - Masanobu Saruta
- Department of Urology, Fujita-Health University School of Medicine, Kutsukake, Toyoake, Aichi Nagoya, Japan
| | - Mamoru Hashimoto
- Department of Urology, Faculty of Medicine, Kindai University, Osakasayama, Osaka, Japan
| | - Takuya Higashio
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Shuya Tsuchida
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Kazuki Nishimura
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Takuya Tsujino
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Ko Nakamura
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Tatsuo Fukushima
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Kyosuke Nishio
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Shutaro Yamamoto
- Department of Urology, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Kosuke Iwatani
- Department of Urology, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Fumihiko Urabe
- Department of Urology, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Keiichiro Mori
- Department of Urology, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Takafumi Yanagisawa
- Department of Urology, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Shunsuke Tsuduki
- Department of Urology, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Kiyoshi Takahara
- Department of Urology, Fujita-Health University School of Medicine, Kutsukake, Toyoake, Aichi Nagoya, Japan
| | - Teruo Inamoto
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Jun Miki
- Department of Urology, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Kazutoshi Fujita
- Department of Urology, Faculty of Medicine, Kindai University, Osakasayama, Osaka, Japan
| | - Takahiro Kimura
- Department of Urology, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan.
| | - Yoshio Ohno
- Department of Urology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Ryoichi Shiroki
- Department of Urology, Fujita-Health University School of Medicine, Kutsukake, Toyoake, Aichi Nagoya, Japan
| | - Hirotsugu Uemura
- Department of Urology, Faculty of Medicine, Kindai University, Osakasayama, Osaka, Japan
| | - Haruhito Azuma
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| |
Collapse
|
200
|
Rajagopal D, MacLeod E, Corogeanu D, Vessillier S. Immune-related adverse events of antibody-based biological medicines in cancer therapy. J Cell Mol Med 2024; 28:e18470. [PMID: 38963257 PMCID: PMC11223167 DOI: 10.1111/jcmm.18470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/03/2024] [Accepted: 05/22/2024] [Indexed: 07/05/2024] Open
Abstract
Recombinant antibodies (Abs) are an integral modality for the treatment of multiple tumour malignancies. Since the Food and Drug Administration (FDA) approval of rituximab as the first monoclonal antibody (mAb) for cancer treatment, several mAbs and antibody (Ab)-based therapies have been approved for the treatment of solid tumour malignancies and other cancers. These Abs function by either blocking oncogenic pathways or angiogenesis, modulating immune response, or by delivering a conjugated drug. The use of Ab-based therapy in cancer patients who could benefit from the treatment, however, is still limited by associated toxicity profiles which may stem from biological features and processes related to target binding, alongside biochemical and/or biophysical characteristics of the therapeutic Ab. A significant immune-related adverse event (irAE) associated with Ab-based therapies is cytokine release syndrome (CRS), characterized by the development of fever, rash and even marked, life-threatening hypotension, and acute inflammation with secondary to systemic uncontrolled increase in a range of pro-inflammatory cytokines. Here, we review irAEs associated with specific classes of approved, Ab-based novel cancer immunotherapeutics, namely immune checkpoint (IC)-targeting Abs, bispecific Abs (BsAbs) and Ab-drug-conjugates (ADCs), highlighting the significance of harmonization in preclinical assay development for safety assessment of Ab-based biotherapeutics as an approach to support and refine clinical translation.
Collapse
Affiliation(s)
- Deepa Rajagopal
- Immunotherapy, Biotherapeutics and Advanced Therapies Division, Science, Research, and Innovation Group, Medicines and Healthcare products Regulatory Agency (MHRA)HertfordshireUK
| | - Elliot MacLeod
- Immunotherapy, Biotherapeutics and Advanced Therapies Division, Science, Research, and Innovation Group, Medicines and Healthcare products Regulatory Agency (MHRA)HertfordshireUK
- Present address:
Gilead Sciences, Winchester HouseOxfordUK
| | - Diana Corogeanu
- Immunotherapy, Biotherapeutics and Advanced Therapies Division, Science, Research, and Innovation Group, Medicines and Healthcare products Regulatory Agency (MHRA)HertfordshireUK
- Present address:
East Sussex Healthcare NHS Trust, Conquest HospitalEast SussexUK
| | - Sandrine Vessillier
- Immunotherapy, Biotherapeutics and Advanced Therapies Division, Science, Research, and Innovation Group, Medicines and Healthcare products Regulatory Agency (MHRA)HertfordshireUK
| |
Collapse
|