51
|
DEGRO practical guideline for central nervous system radiation necrosis part 2: treatment. Strahlenther Onkol 2022; 198:971-980. [PMID: 36038670 PMCID: PMC9581806 DOI: 10.1007/s00066-022-01973-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 06/15/2022] [Indexed: 11/08/2022]
Abstract
Purpose The Working Group for Neurooncology of the German Society for Radiation Oncology (DEGRO; AG NRO) in cooperation with members of the Neurooncological Working Group of the German Cancer Society (DKG-NOA) aimed to define a practical guideline for the diagnosis and treatment of radiation-induced necrosis (RN) of the central nervous system (CNS). Methods Panel members of the DEGRO working group invited experts, participated in a series of conferences, supplemented their clinical experience, performed a literature review, and formulated recommendations for medical treatment of RN, including bevacizumab, in clinical routine. Conclusion Diagnosis and treatment of RN requires multidisciplinary structures of care and defined processes. Diagnosis has to be made on an interdisciplinary level with the joint knowledge of a neuroradiologist, radiation oncologist, neurosurgeon, neuropathologist, and neurooncologist. If the diagnosis of blood–brain barrier disruptions (BBD) or RN is likely, treatment should be initiated depending on the symptoms, location, and dynamic of the lesion. Multiple treatment options are available (such as observation, surgery, steroids, and bevacizumab) and the optimal approach should be discussed in an interdisciplinary setting. In this practice guideline, we offer detailed treatment strategies for various scenarios.
Collapse
|
52
|
Immunotherapy for Aggressive and Metastatic Pituitary Neuroendocrine Tumors (PitNETs): State-of-the Art. Cancers (Basel) 2022; 14:cancers14174093. [PMID: 36077631 PMCID: PMC9454884 DOI: 10.3390/cancers14174093] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/20/2022] [Accepted: 08/21/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Aggressive and metastatic PitNETs are challenging conditions. Immune checkpoint inhibitors (ICIs) are currently considered in cases resistant to temozolomide (TMZ). However, clinical experience is essentially limited to case reports, with variable outcomes. Material and Methods: The effects of ICIs on 12 aggressive/metastatic PitNETs from the literature were reviewed and analyzed according to tumor characteristics, with the additional description of a silent-Pit1 metastatic tumor responding to pembrolizumab. Results: Most cases were metastatic (10/13: 6 corticotroph, 3 lactotroph, 1 silent Pit1); 3 were aggressive (2 corticotroph, 1 lactotroph). ICIS was used either as monotherapy or in combination. At last follow-up on ICI, a complete response (CR) was present in 3 cases and a partial response (PR) in 2 cases (4/5 metastatic). One sustained stable disease (SD) was reported. Progressive disease (PD) was observed in 7 cases, 3 of them after initial SD (n = 1) or PR (n = 3), with 2 reported deaths. PDL1 expression was studied in 10 cases and was high (>95%) in 2 Pit1-derived metastatic PitNETs (1 CR and 1 remarkable PR) but absent/low (<1%) in the remaining cases (including 1 CP and 2 PR). Elevated tumor mutation burden could be informative in corticotroph PitNETs, especially in mismatch repair-deficient tumors. Conclusion: Significant benefits from ICIs were documented in about half of TMZ-resistant PitNETS. High PDL1 expression was associated with remarkable responses but may be dispensable. Based on their acceptable tolerance and awaiting recognized predictors of response, ICIs may be considered a valuable option for such patients.
Collapse
|
53
|
Persson ML, Douglas AM, Alvaro F, Faridi P, Larsen MR, Alonso MM, Vitanza NA, Dun MD. The intrinsic and microenvironmental features of diffuse midline glioma; implications for the development of effective immunotherapeutic treatment strategies. Neuro Oncol 2022; 24:1408-1422. [PMID: 35481923 PMCID: PMC9435509 DOI: 10.1093/neuonc/noac117] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Diffuse midline glioma (DMG), including those of the brainstem (diffuse intrinsic pontine glioma), are pediatric tumors of the central nervous system (CNS). Recognized as the most lethal of all childhood cancers, palliative radiotherapy remains the only proven treatment option, however, even for those that respond, survival is only temporarily extended. DMG harbor an immunologically “cold” tumor microenvironment (TME) with few infiltrating immune cells. The mechanisms underpinning the cold TME are not well understood. Low expression levels of immune checkpoint proteins, including PD-1, PD-L1, and CTLA-4, are recurring features of DMG and likely contribute to the lack of response to immune checkpoint inhibitors (ICIs). The unique epigenetic signatures (including stem cell-like methylation patterns), a low tumor mutational burden, and recurring somatic mutations (H3K27M, TP53, ACVR1, MYC, and PIK3CA), possibly play a role in the reduced efficacy of traditional immunotherapies. Therefore, to circumvent the lack of efficacy thus far seen for the use of ICIs, adoptive cell transfer (including chimeric antigen receptor T cells) and the use of oncolytic viruses, are currently being evaluated for the treatment of DMG. It remains an absolute imperative that we improve our understanding of DMG’s intrinsic and TME features if patients are to realize the potential benefits offered by these sophisticated treatments. Herein, we summarize the limitations of immunotherapeutic approaches, highlight the emerging safety and clinical efficacy shown for sophisticated cell-based therapies, as well as the evolving knowledge underpinning the DMG-immune axis, to guide the development of immunotherapies that we hope will improve outcomes.
Collapse
Affiliation(s)
- Mika L Persson
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia.,Precision Medicine Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Alicia M Douglas
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia.,Precision Medicine Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Frank Alvaro
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia.,Precision Medicine Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,John Hunter Children's Hospital, New Lambton Heights, NSW, Australia
| | - Pouya Faridi
- Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, VIC, Australia
| | - Martin R Larsen
- Department of Molecular Biology and Biochemistry, Protein Research Group, University of Southern Denmark, Odense, Denmark
| | - Marta M Alonso
- Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for Applied Medical Research (CIMA), Pamplona, Spain
| | - Nicholas A Vitanza
- The Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA.,Division of Pediatric Hematology, Oncology, Bone Marrow Transplant, and Cellular Therapy, Department of Pediatrics, Seattle Children's Hospital, Seattle, WA, USA
| | - Matthew D Dun
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia.,Precision Medicine Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
54
|
Yennurajalingam S, Bruera E. Do Patients Benefit from a Trial of Corticosteroids at the End of Life? Curr Treat Options Oncol 2022; 23:796-805. [PMID: 35362799 DOI: 10.1007/s11864-022-00977-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2022] [Indexed: 11/03/2022]
Abstract
OPINION STATEMENT Patients with advanced cancer in the last 6 months of their lives have a higher frequency of distressing and debilitating physical and psychosocial symptoms such as cancer pain, cancer-related fatigue (CRF), anorexia, shortness of breath, poor sleep, anxiety, and depression. Often these symptoms significantly impact the patients' quality of life, and therefore require prompt assessment and effective treatment. There are specific treatments for certain distressing cancer-related symptoms (e.g., opioids for pain), but for the other symptoms such as CRF, anorexia-cachexia, and shortness of breath, there are limited or no evidence-based treatments. Also, in the management of cancer pain in this population, many patients are refractory to opioids. Hence, corticosteroids are one of the most common adjuvant medications prescribed for the management of this distressing symptom. However, there is limited evidence in regard to the effectiveness of corticosteroids in the improvement of the symptoms, side-effect profile, most optimal duration of use, dose, type of steroid, and most recently, the use with immunotherapy in advanced cancer patients at the end of life. These factors significantly limit the use of this important medication in terminally ill cancer patients. Further research is therefore critical to provide the optimal prescription of corticosteroids in this highly distressed population.
Collapse
Affiliation(s)
- Sriram Yennurajalingam
- Department of Palliative Care Rehabilitation, and Integrative Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. #1414, Houston, TX, 77030, USA.
| | - Eduardo Bruera
- Department of Palliative Care Rehabilitation, and Integrative Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. #1414, Houston, TX, 77030, USA
| |
Collapse
|
55
|
Neurotoxicity and safety of the rechallenge of immune checkpoint inhibitors: a growing issue in neuro-oncology practice. Neurol Sci 2022; 43:2339-2361. [DOI: 10.1007/s10072-022-05920-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/19/2022] [Indexed: 10/19/2022]
|
56
|
Kalfeist L, Galland L, Ledys F, Ghiringhelli F, Limagne E, Ladoire S. Impact of Glucocorticoid Use in Oncology in the Immunotherapy Era. Cells 2022; 11:770. [PMID: 35269392 PMCID: PMC8909189 DOI: 10.3390/cells11050770] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/18/2022] [Accepted: 02/18/2022] [Indexed: 12/11/2022] Open
Abstract
Thanks to their anti-inflammatory, anti-oedema, and anti-allergy properties, glucocorticoids are among the most widely prescribed drugs in patients with cancer. The indications for glucocorticoid use are very wide and varied in the context of cancer and include the symptomatic management of cancer-related symptoms (compression, pain, oedema, altered general state) but also prevention or treatment of common side effects of anti-cancer therapies (nausea, allergies, etc.) or immune-related adverse events (irAE). In this review, we first give an overview of the different clinical situations where glucocorticoids are used in oncology. Next, we describe the current state of knowledge regarding the effects of these molecules on immune response, in particular anti-tumour response, and we summarize available data evaluating how these effects may interfere with the efficacy of immunotherapy using immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Laura Kalfeist
- Platform of Transfer in Cancer Biology, Georges-Francois Leclerc Center, 21000 Dijon, France; (L.K.); (L.G.); (F.L.); (F.G.); (E.L.)
- UMR INSERM 1231 “Lipides Nutrition Cancer”, 21000 Dijon, France
| | - Loïck Galland
- Platform of Transfer in Cancer Biology, Georges-Francois Leclerc Center, 21000 Dijon, France; (L.K.); (L.G.); (F.L.); (F.G.); (E.L.)
- UMR INSERM 1231 “Lipides Nutrition Cancer”, 21000 Dijon, France
- Department of Medical Oncology, Georges-François Leclerc Center, 21000 Dijon, France
| | - Fanny Ledys
- Platform of Transfer in Cancer Biology, Georges-Francois Leclerc Center, 21000 Dijon, France; (L.K.); (L.G.); (F.L.); (F.G.); (E.L.)
- UMR INSERM 1231 “Lipides Nutrition Cancer”, 21000 Dijon, France
| | - François Ghiringhelli
- Platform of Transfer in Cancer Biology, Georges-Francois Leclerc Center, 21000 Dijon, France; (L.K.); (L.G.); (F.L.); (F.G.); (E.L.)
- UMR INSERM 1231 “Lipides Nutrition Cancer”, 21000 Dijon, France
- Department of Medical Oncology, Georges-François Leclerc Center, 21000 Dijon, France
- School of Medicine, University of Burgundy Franche-Comté, 21000 Dijon, France
| | - Emeric Limagne
- Platform of Transfer in Cancer Biology, Georges-Francois Leclerc Center, 21000 Dijon, France; (L.K.); (L.G.); (F.L.); (F.G.); (E.L.)
- UMR INSERM 1231 “Lipides Nutrition Cancer”, 21000 Dijon, France
| | - Sylvain Ladoire
- Platform of Transfer in Cancer Biology, Georges-Francois Leclerc Center, 21000 Dijon, France; (L.K.); (L.G.); (F.L.); (F.G.); (E.L.)
- UMR INSERM 1231 “Lipides Nutrition Cancer”, 21000 Dijon, France
- Department of Medical Oncology, Georges-François Leclerc Center, 21000 Dijon, France
- School of Medicine, University of Burgundy Franche-Comté, 21000 Dijon, France
| |
Collapse
|
57
|
Liu CY, Li C. Clinical Features and Risk Factors of Severe Pneumonia in Children With Acute Lymphoblastic Leukemia. Front Pediatr 2022; 10:813638. [PMID: 35601429 PMCID: PMC9120655 DOI: 10.3389/fped.2022.813638] [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: 11/12/2021] [Accepted: 03/23/2022] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE This study aims to analyze the clinical characteristics of pediatric acute lymphoblastic leukemia (ALL) complicated by pneumonia and the risk factors of severe cases to preliminarily construct a prediction model for ALL complicated by severe pneumonia. METHODS A retrospective analysis was carried out on the clinical data of children diagnosed with ALL complicated by pneumonia hospitalized at the Department of Pediatrics of the Affiliated Hospital of Southwest Medical University between January 2013 and December 2020. The risk factors of severe ALL complicated by pneumonia were investigated with logistic regression analysis, and the risk prediction model was constructed. RESULTS A total of 116 cases of pediatric ALL complicated by pneumonia were analyzed. There were 71 cases of mild pneumonia and 45 cases of severe pneumonia. The main clinical manifestations were cough in 112 cases and fever in 109. Pathogens were detected in 23 cases. Multiple regression factor analysis indicated that the use of hormones (OR 4.001, 95% CI: 1.505-10.632), neutropenia or agranulocytosis (OR 7.472, 95% CI: 2.710-20.602), hemoglobin (Hb) < 90 g/L (OR 3.270, 95% CI: 1.256~8.516), and C-reactive protein (CRP) >15 mg/L (OR 3.253, 95% CI: 1.209~8.751) were independent risk factors that were associated with severe pneumonia. Logistic regression was used to establish the risk prediction model of ALL with severe pneumonia. The p-value was 0.659. The area under the receiver operating characteristic curve was 0.851, and the sensitivity and specificity were 84.4 and 71.8%, respectively. CONCLUSION The development of severe pneumonia may be affected by the use of hormones, neutropenia or agranulocytosis, Hb < 90 g/L, and CRP > 15 mg/L. The prediction model based on the risk factors is effective, which can provide a reference for the clinical evaluation of acute lymphoblastic leukemia with severe pneumonia.
Collapse
Affiliation(s)
- Chun-Yan Liu
- Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, Luzhou, China
| | - Cheng Li
- Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, Luzhou, China
| |
Collapse
|
58
|
Arai K, Matsuda M, Nakayasu H, Meguro S, Kurokami T, Kubota A, Iwasaki T, Suzuki M, Kawaguchi S, Iwashita T. Nivolumab-induced liver injury with a steroid-refractory increase in biliary enzymes, in a patient with malignant mesothelioma: An autopsy case report. Clin Case Rep 2021; 9:e05174. [PMID: 34987810 PMCID: PMC8697699 DOI: 10.1002/ccr3.5174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/29/2021] [Accepted: 11/07/2021] [Indexed: 12/19/2022] Open
Abstract
This is the first autopsy report of hepatotoxicity from nivolumab immunotherapy for malignant mesothelioma. The increase in levels of biliary enzymes and randomly distributed endothelial damage were steroid-refractory, but second-line option was abandoned because of cachexia. Further discussions are needed regarding the customized management of immune-related toxicities.
Collapse
Affiliation(s)
- Kazumori Arai
- Department of PathologyShizuoka General HospitalShizuokaJapan
| | - Masanori Matsuda
- Department of GastroenterologyShizuoka General HospitalShizuokaJapan
| | - Hiromasa Nakayasu
- Department of Respiratory MedicineShizuoka General HospitalShizuokaJapan
| | - Shiori Meguro
- Department of Regenerative and Infectious PathologyHamamatsu University School of MedicineHamamatsuJapan
| | - Takafumi Kurokami
- Department of GastroenterologyShizuoka General HospitalShizuokaJapan
| | - Aki Kubota
- Department of PathologyShizuoka General HospitalShizuokaJapan
| | | | - Makoto Suzuki
- Department of PathologyShizuoka General HospitalShizuokaJapan
| | - Shinya Kawaguchi
- Department of GastroenterologyShizuoka General HospitalShizuokaJapan
| | - Toshihide Iwashita
- Department of Regenerative and Infectious PathologyHamamatsu University School of MedicineHamamatsuJapan
| |
Collapse
|
59
|
Fediw M, Lau K. Novel Cancer Therapeutics and Implications for Rehabilitation. CURRENT PHYSICAL MEDICINE AND REHABILITATION REPORTS 2021. [DOI: 10.1007/s40141-021-00323-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
60
|
Montisci A, Vietri MT, Palmieri V, Sala S, Donatelli F, Napoli C. Cardiac Toxicity Associated with Cancer Immunotherapy and Biological Drugs. Cancers (Basel) 2021; 13:4797. [PMID: 34638281 PMCID: PMC8508330 DOI: 10.3390/cancers13194797] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 12/19/2022] Open
Abstract
Cancer immunotherapy significantly contributed to an improvement in the prognosis of cancer patients. Immunotherapy, including human epidermal growth factor receptor 2 (HER2)-targeted therapies, immune checkpoint inhibitors (ICI), and chimeric antigen receptor-modified T (CAR-T), share the characteristic to exploit the capabilities of the immune system to kill cancerous cells. Trastuzumab is a monoclonal antibody against HER2 that prevents HER2-mediated signaling; it is administered mainly in HER2-positive cancers, such as breast, colorectal, biliary tract, and non-small-cell lung cancers. Immune checkpoint inhibitors (ICI) inhibit the binding of CTLA-4 or PD-1 to PDL-1, allowing T cells to kill cancerous cells. ICI can be used in melanomas, non-small-cell lung cancer, urothelial, and head and neck cancer. There are two main types of T-cell transfer therapy: tumor-infiltrating lymphocytes (or TIL) therapy and chimeric antigen receptor-modified T (CAR-T) cell therapy, mainly applied for B-cell lymphoma and leukemia and mantle-cell lymphoma. HER2-targeted therapies, mainly trastuzumab, are associated with left ventricular dysfunction, usually reversible and rarely life-threatening. PD/PDL-1 inhibitors can cause myocarditis, rare but potentially fulminant and associated with a high fatality rate. CAR-T therapy is associated with several cardiac toxic effects, mainly in the context of a systemic adverse effect, the cytokines release syndrome.
Collapse
Affiliation(s)
- Andrea Montisci
- Division of Cardiothoracic Intensive Care, ASST Spedali Civili, 25123 Brescia, Italy;
| | - Maria Teresa Vietri
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 81100 Naples, Italy;
| | - Vittorio Palmieri
- Department of Cardiac Surgery and Transplantation, Ospedali dei Colli Monaldi-Cotugno-CTO, 80131 Naples, Italy;
| | - Silvia Sala
- Department of Anesthesia and Intensive Care, University of Brescia, 25121 Brescia, Italy;
| | - Francesco Donatelli
- Cardiac Surgery, University of Milan, 20122 Milan, Italy
- Department of Cardiac Surgery, Istituto Clinico Sant’Ambrogio, 20149 Milan, Italy
| | - Claudio Napoli
- Clinical Department of Internal Medicine and Specialistics, University Department of Advanced Clinical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 81100 Naples, Italy;
- IRCCS SDN, 80143 Naples, Italy
| |
Collapse
|
61
|
Paderi A, Gambale E, Botteri C, Giorgione R, Lavacchi D, Brugia M, Mazzoni F, Giommoni E, Bormioli S, Amedei A, Pillozzi S, Matucci Cerinic M, Antonuzzo L. Association of Systemic Steroid Treatment and Outcome in Patients Treated with Immune Checkpoint Inhibitors: A Real-World Analysis. Molecules 2021; 26:5789. [PMID: 34641331 PMCID: PMC8510096 DOI: 10.3390/molecules26195789] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/15/2021] [Accepted: 09/15/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Immune-related adverse events (irAEs) are inflammatory side effects, which can occur during immune-checkpoint(s) inhibitors (ICIs) therapy. Steroids are the first-line agents to manage irAEs because of their immunosuppressive properties. However, it is still debated whether or when steroids can be administered without abrogating the therapeutic efforts of immunotherapy. METHODS We retrospectively evaluated 146 patients with metastatic non-small cell lung cancer (NSCLC), melanoma and renal cell carcinoma (RCC) treated with ICIs. We assessed the progression-free survival (PFS) of patients treated with steroids due to an irAE compared to a no-steroid group. RESULTS The early treatment with steroid (within the first 30 days from the beginning of immunotherapy) was not related to a shorter PFS (p = 0.077). Interestingly, patients who were treated with steroids after 30 days from the start of immunotherapy had significantly longer PFS (p = 0.017). In a multivariate analysis, treatment with steroids after 30 days was an independent prognostic factor for PFS (HR: 0.59 [95% CI 0.36-0.97], p = 0.037). CONCLUSIONS This retrospective study points out that early systemic steroids administration to manage irAEs might not have a detrimental effect on patient clinical outcome in NSCLC, melanoma and RCC patients.
Collapse
Affiliation(s)
- Agnese Paderi
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Elisabetta Gambale
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Cristina Botteri
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Roberta Giorgione
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Daniele Lavacchi
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Marco Brugia
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Francesca Mazzoni
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Elisa Giommoni
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Susanna Bormioli
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (S.B.); (A.A.); (M.M.C.)
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (S.B.); (A.A.); (M.M.C.)
| | - Serena Pillozzi
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
| | - Marco Matucci Cerinic
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (S.B.); (A.A.); (M.M.C.)
| | - Lorenzo Antonuzzo
- Medical Oncology Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy; (A.P.); (E.G.); (C.B.); (R.G.); (D.L.); (M.B.); (F.M.); (E.G.); (S.P.)
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (S.B.); (A.A.); (M.M.C.)
| |
Collapse
|
62
|
Firer MA, Shapira MY, Luboshits G. The Impact of Induction Regimes on Immune Responses in Patients with Multiple Myeloma. Cancers (Basel) 2021; 13:4090. [PMID: 34439244 PMCID: PMC8393868 DOI: 10.3390/cancers13164090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/16/2022] Open
Abstract
Current standard frontline therapy for newly diagnosed patients with multiple myeloma (NDMM) involves induction therapy, autologous stem cell transplantation (ASCT), and maintenance therapy. Major efforts are underway to understand the biological and the clinical impacts of each stage of the treatment protocols on overall survival statistics. The most routinely used drugs in the pre-ASCT "induction" regime have different mechanisms of action and are employed either as monotherapies or in various combinations. Aside from their direct effects on cancer cell mortality, these drugs are also known to have varying effects on immune cell functionality. The question remains as to how induction therapy impacts post-ASCT immune reconstitution and anti-tumor immune responses. This review provides an update on the known immune effects of melphalan, dexamethasone, lenalidomide, and bortezomib commonly used in the induction phase of MM therapy. By analyzing the actions of each individual drug on the immune system, we suggest it might be possible to leverage their effects to rationally devise more effective induction regimes. Given the genetic heterogeneity between myeloma patients, it may also be possible to identify subgroups of patients for whom particular induction drug combinations would be more appropriate.
Collapse
Affiliation(s)
- Michael A. Firer
- Department Chemical Engineering, Ariel University, Ariel 40700, Israel;
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel
- Ariel Center for Applied Cancer Research, Ariel University, Ariel 40700, Israel
| | - Michael Y. Shapira
- The Hematology Institute, Assuta Medical Center, Tel Aviv 6971028, Israel;
| | - Galia Luboshits
- Department Chemical Engineering, Ariel University, Ariel 40700, Israel;
- Ariel Center for Applied Cancer Research, Ariel University, Ariel 40700, Israel
| |
Collapse
|
63
|
Miles D, Gligorov J, André F, Cameron D, Schneeweiss A, Barrios C, Xu B, Wardley A, Kaen D, Andrade L, Semiglazov V, Reinisch M, Patel S, Patre M, Morales L, Patel SL, Kaul M, Barata T, O'Shaughnessy J. Primary results from IMpassion131, a double-blind, placebo-controlled, randomised phase III trial of first-line paclitaxel with or without atezolizumab for unresectable locally advanced/metastatic triple-negative breast cancer. Ann Oncol 2021; 32:994-1004. [PMID: 34219000 DOI: 10.1016/j.annonc.2021.05.801] [Citation(s) in RCA: 421] [Impact Index Per Article: 105.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND In the phase III IMpassion130 trial, combining atezolizumab with first-line nanoparticle albumin-bound-paclitaxel for advanced triple-negative breast cancer (aTNBC) showed a statistically significant progression-free survival (PFS) benefit in the intention-to-treat (ITT) and programmed death-ligand 1 (PD-L1)-positive populations, and a clinically meaningful overall survival (OS) effect in PD-L1-positive aTNBC. The phase III KEYNOTE-355 trial adding pembrolizumab to chemotherapy for aTNBC showed similar PFS effects. IMpassion131 evaluated first-line atezolizumab-paclitaxel in aTNBC. PATIENTS AND METHODS Eligible patients [no prior systemic therapy or ≥12 months since (neo)adjuvant chemotherapy] were randomised 2:1 to atezolizumab 840 mg or placebo (days 1, 15), both with paclitaxel 90 mg/m2 (days 1, 8, 15), every 28 days until disease progression or unacceptable toxicity. Stratification factors were tumour PD-L1 status, prior taxane, liver metastases and geographical region. The primary endpoint was investigator-assessed PFS, tested hierarchically first in the PD-L1-positive [immune cell expression ≥1%, VENTANA PD-L1 (SP142) assay] population, and then in the ITT population. OS was a secondary endpoint. RESULTS Of 651 randomised patients, 45% had PD-L1-positive aTNBC. At the primary PFS analysis, adding atezolizumab to paclitaxel did not improve investigator-assessed PFS in the PD-L1-positive population [hazard ratio (HR) 0.82, 95% confidence interval (CI) 0.60-1.12; P = 0.20; median PFS 6.0 months with atezolizumab-paclitaxel versus 5.7 months with placebo-paclitaxel]. In the PD-L1-positive population, atezolizumab-paclitaxel was associated with more favourable unconfirmed best overall response rate (63% versus 55% with placebo-paclitaxel) and median duration of response (7.2 versus 5.5 months, respectively). Final OS results showed no difference between arms (HR 1.11, 95% CI 0.76-1.64; median 22.1 months with atezolizumab-paclitaxel versus 28.3 months with placebo-paclitaxel in the PD-L1-positive population). Results in the ITT population were consistent with the PD-L1-positive population. The safety profile was consistent with known effects of each study drug. CONCLUSION Combining atezolizumab with paclitaxel did not improve PFS or OS versus paclitaxel alone. CLINICALTRIALS.GOV: NCT03125902.
Collapse
Affiliation(s)
- D Miles
- Mount Vernon Cancer Centre, Northwood, UK.
| | - J Gligorov
- Medical Oncology Department, Institut Universitaire de Cancérologie Assistance Publique-Hôpitaux de Paris-Sorbonne Université, Paris, France
| | - F André
- Department of Medical Oncology, Gustave Roussy, Université Paris Sud, Villejuif, France
| | - D Cameron
- University of Edinburgh, Edinburgh, UK
| | - A Schneeweiss
- Division of Gynecologic Oncology, National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - C Barrios
- Latin American Cooperative Oncology Group, Porto Alegre RS, Brazil
| | - B Xu
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - A Wardley
- National Institute for Health Research Manchester Clinical Research Facility at The Christie NHS Foundation Trust, Manchester, UK; Outreach Research & Innovation Group, Manchester, UK
| | - D Kaen
- Centro Oncológico Riojano Integral and Universidad Nacional de La Rioja, La Rioja, Argentina
| | - L Andrade
- Clinical Oncology, Santa Casa de Misericórdia da Bahia, Salvador, Brazil
| | - V Semiglazov
- NN Petrov Research Institute of Oncology, St. Petersburg, Russia
| | | | - S Patel
- Product Development Oncology, Genentech, Inc., South San Francisco, USA
| | - M Patre
- Global Product Development Medical Affairs Oncology, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - L Morales
- Global Product Development Medical Affairs Oncology, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - S L Patel
- Patient-Centered Outcomes Research, Genentech, Inc., South San Francisco, USA
| | - M Kaul
- Product Development Oncology, Genentech, Inc., South San Francisco, USA
| | - T Barata
- Pharma Development Biostatistics Oncology, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - J O'Shaughnessy
- Baylor University Medical Center, Texas Oncology, US Oncology, Dallas, USA
| |
Collapse
|
64
|
Zheng JY, Mixon AC, McLarney MD. Safety, Precautions, and Modalities in Cancer Rehabilitation: an Updated Review. CURRENT PHYSICAL MEDICINE AND REHABILITATION REPORTS 2021; 9:142-153. [PMID: 34178432 PMCID: PMC8214054 DOI: 10.1007/s40141-021-00312-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Providing rehabilitation to patients with cancer can be challenging due to the medical complexity associated with the illness and its treatments. This article provides the reader with a summary of frequently encountered medical conditions in the cancer population and associated safety considerations and precautions. An update on treatment modalities commonly used for symptom management is also presented. RECENT FINDINGS Cancer and cancer treatments can cause changes in multiple organ systems. Special considerations and precautions are necessary to provide safe and effective rehabilitation. Physical modalities can be used as monotherapy or adjunct to treatment for common cancer-related side effects with recent studies noting benefit with a variety of modalities. SUMMARY Detailed assessment of the cancer patient is necessary before implementing a rehabilitation program. Understanding cancer and side effects of treatments, including newer options, are necessary to provide safe care.
Collapse
Affiliation(s)
- Jasmine Y. Zheng
- Department of Physical Medicine and Rehabilitation, University of Pennsylvania, 1800 Lombard St, 1st Floor, Philadelphia, PA 19146 USA
| | - Alyssa C. Mixon
- Department of Physical Medicine and Rehabilitation, University of Pennsylvania, 1800 Lombard St, 1st Floor, Philadelphia, PA 19146 USA
| | - Mitra D. McLarney
- Department of Physical Medicine and Rehabilitation, University of Pennsylvania, 1800 Lombard St, 1st Floor, Philadelphia, PA 19146 USA
| |
Collapse
|
65
|
[Impact of the use of systemic corticosteroid therapy on the effectiveness of immune checkpoint inhibitors]. Bull Cancer 2021; 108:635-642. [PMID: 33888298 DOI: 10.1016/j.bulcan.2021.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/16/2021] [Accepted: 01/20/2021] [Indexed: 12/20/2022]
Abstract
Immunotherapy, which consists in using molecules targeting the immune system, has existed for many years in oncology (vaccines, interleukins, monoclonal antibodies) but has recently expanded due to the development of immune checkpoint inhibitors. These monoclonal antibodies help to restore the immunity against cancer by specifically targeting some immune checkpoints such as CTLA-4, PD-1 and PD-L1. Furthermore, in oncology, it is common to use systemic corticosteroids in the management of symptoms linked to the natural history of the disease (pain, spinal cord compression, cerebral edema) and toxicities linked to anticancer treatment. The impact of corticosteroids on the efficacy of immune checkpoint inhibitors is still poorly understood and they should be used cautiously. According to previously published studies, there seems to be a deleterious effect of corticosteroid therapy on the efficacy of immune checkpoint inhibitors when administered before or at the initiation of immunotherapy, while this effect does not seem present when corticosteroids are administered to patients already undergoing immunotherapy. The aim of this work is to analyze the existing data evaluating the impact of corticosteroid use of on the efficacy of immune checkpoint inhibitors.
Collapse
|