1
|
Mielczarek Ł, Brodziak A, Sobczuk P, Kawecki M, Cudnoch-Jędrzejewska A, Czarnecka AM. Renal toxicity of targeted therapies for renal cell carcinoma in patients with normal and impaired kidney function. Cancer Chemother Pharmacol 2021; 87:723-742. [PMID: 33768301 PMCID: PMC8110505 DOI: 10.1007/s00280-021-04260-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 03/13/2021] [Indexed: 12/11/2022]
Abstract
The introduction of novel targeted therapies during the last 2 decades has led to a significant improvement in patients' clinical outcomes with renal cell carcinoma. However, this improvement came at the price of a whole new spectrum of adverse events, including renal toxicity. Systemic treatment of patients with kidney neoplasms who often present with impairment of kidney function, even prior to treatment, poses an increasing diagnostic and therapeutic challenge for clinicians. Common lifestyle-related comorbidities, i.e., hypertension and diabetes, may contribute to further impairment of kidney function. The lack of official guidelines and the exclusion of patients with reduced kidney function from the clinical trials of recently approved drugs complicate the issue even further. Early detection and correct management of renal toxic effects are crucial to preserve kidney function and ensure the optimal administration of life-prolonging therapies. This review presents detailed information on the renal toxicities of three groups of drugs commonly used in renal cell carcinoma treatment: tyrosine kinase inhibitors, mammalian target of rapamycin inhibitors, and immune checkpoint inhibitors. We outline the incidence and underlying mechanisms of renal adverse effects with a focus on patients on renal replacement therapy, as well as present suggestions for their management.
Collapse
Affiliation(s)
- Łukasz Mielczarek
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Anna Brodziak
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
- Department of Oncology and Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Paweł Sobczuk
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Maciej Kawecki
- Department of Oncology and Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Anna M Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
2
|
Thoracic Manifestations of Genitourinary Neoplasms and Treatment-related Complications. J Thorac Imaging 2019; 34:W36-W48. [PMID: 31009398 DOI: 10.1097/rti.0000000000000382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Genitourinary (GU) malignancies are a diverse group of common and uncommon neoplasms that may be associated with significant mortality. Metastases from GU neoplasms are frequently encountered in the chest, and virtually all thoracic structures can be involved. Although the most common imaging manifestations include hematogenous dissemination manifesting with peripheral predominant bilateral pulmonary nodules and lymphatic metastases manifesting with mediastinal and hilar lymphadenopathy, some GU malignancies exhibit unique features. We review the general patterns, pathways, and thoracic imaging features of renal, adrenal, urothelial, prostatic, and testicular metastatic neoplasms, as well as provide a discussion of treatment-related complications that might manifest in the chest. Detailed reporting of these patterns will allow the imager to assist the referring clinicians and surgeons in accurate determination of the stage, prognosis, and treatment options available for the patient. Awareness of specific treatment-related complications further allows the imager to enhance patient safety through accurate and timely reporting of potentially life-threatening consequences of therapies.
Collapse
|
3
|
Méndez-Vidal MJ, Molina Á, Anido U, Chirivella I, Etxaniz O, Fernández-Parra E, Guix M, Hernández C, Lambea J, Montesa Á, Pinto Á, Ros S, Gallardo E. Pazopanib: Evidence review and clinical practice in the management of advanced renal cell carcinoma. BMC Pharmacol Toxicol 2018; 19:77. [PMID: 30477570 PMCID: PMC6258404 DOI: 10.1186/s40360-018-0264-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/29/2018] [Indexed: 12/19/2022] Open
Abstract
Background Pazopanib is indicated in the first-line treatment of metastatic renal cell cancer (mRCC). The aim of this study was to review the efficacy, safety, and pharmacokinetics of pazopanib and see how these aspects are linked to clinical practice. Methods A non-exhaustive systematic review was conducted according to the three topics. No publication restrictions were imposed and the selected languages were Spanish and English. After that, a summary of the main results and findings of the review was presented and discussed during three meetings (one for each topic) with 13 medical oncologists that usually treat mRCC. At these meetings, a questionnaire on the first-line use of pazopanib in clinical practice was also drawn up. After the meetings, the questionnaire was completed by 60 specialist medical oncologists in renal cancer. Results The efficacy and safety of pazopanib have been demonstrated in several clinical trials, and subsequently confirmed in studies in real-world clinical practice. In addition to its clinical benefit and good safety profile, quality of life results for pazopanib, which compare favorably to sunitinib, make it a good option in the first-line treatment of patients. Special populations have been included in studies conducted with pazopanib, and it is safe for use in elderly patients, poor functional status, kidney failure, and mild or moderate hepatic impairment, and in patients with concomitant cardiovascular disease. The results of the questionnaire have shown that pazopanib is perceived as an effective drug, in which quality of life (QoL) outcomes are valued above all. Conclusions This paper offers a comprehensive and critical summary of efficacy, tolerability, and pharmacokinetics of pazopanib in the treatment of mRCC. Pazopanib is an effective treatment with an acceptable safety profile. Its QoL and tolerability results offer certain advantages when compared with other therapeutic alternatives, and its use appears to be safe in different patient profiles.
Collapse
Affiliation(s)
- María José Méndez-Vidal
- Oncology Department, Maimonides Institute of Biomedical Research (IMIBIC), Reina Sofia Hospital, Córdoba, Spain
| | - Áurea Molina
- Oncology Department, Complejo Hospitalario Universitario A Coruña, ACoruña, Spain
| | - Urbano Anido
- Oncology Department, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, Spain
| | - Isabel Chirivella
- Oncology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Olatz Etxaniz
- Oncology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | | | - Marta Guix
- Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Carolina Hernández
- Medical Oncology Department, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Julio Lambea
- Medical Oncology Department, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - Álvaro Montesa
- Medical Oncology Department, Hospital Regional de Málaga, Málaga, Spain
| | - Álvaro Pinto
- Medical Oncology Department, Hospital la Paz, Madrid, Spain
| | - Silverio Ros
- Oncology Department, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Enrique Gallardo
- Oncology Department, Parc Taulí Hospital Universitari. Institut d'Investigació i Innovació Parc Taulí I3PT. Universitat Autònoma de Barcelona, Sabadell, Barcelona, Spain.
| |
Collapse
|
4
|
Implementation of a Nurse-driven Educational Program Improves Management of Sorafenib’s Toxicities in Hepatocellular Carcinoma. Cancer Nurs 2018; 41:418-423. [DOI: 10.1097/ncc.0000000000000521] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
5
|
Dong J, Zhang Q, Wang Z, Huang G, Li S. Recent Advances in the Development of Indazole-based Anticancer Agents. ChemMedChem 2018; 13:1490-1507. [PMID: 29863292 DOI: 10.1002/cmdc.201800253] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/25/2018] [Indexed: 12/20/2022]
Abstract
Cancer is one of the leading causes of human mortality globally; therefore, intensive efforts have been made to seek new active drugs with improved anticancer efficacy. Indazole-containing derivatives are endowed with a broad range of biological properties, including anti-inflammatory, antimicrobial, anti-HIV, antihypertensive, and anticancer activities. In recent years, the development of anticancer drugs has given rise to a wide range of indazole derivatives, some of which exhibit outstanding activity against various tumor types. The aim of this review is to outline recent developments concerning the anticancer activity of indazole derivatives, as well as to summarize the design strategies and structure-activity relationships of these compounds.
Collapse
Affiliation(s)
- Jinyun Dong
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, China
| | - Qijing Zhang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, China
| | - Zengtao Wang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, China
| | - Guang Huang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, China
| | - Shaoshun Li
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, China
| |
Collapse
|
6
|
Tannir NM, Pal SK, Atkins MB. Second-Line Treatment Landscape for Renal Cell Carcinoma: A Comprehensive Review. Oncologist 2018; 23:540-555. [PMID: 29487224 DOI: 10.1634/theoncologist.2017-0534] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/03/2018] [Indexed: 12/12/2022] Open
Abstract
The management of advanced clear-cell renal cell carcinoma has steadily improved over the past decade with the introduction of antiangiogenic and targeted therapies. Recently, three new therapies have been approved for use as second-line options that further advance the treatment armamentarium: nivolumab, a monoclonal antibody targeting the programmed cell death receptor; cabozantinib, a small-molecule tyrosine kinase inhibitor (TKI) of vascular endothelial growth factor receptor (VEGFR), MET, and AXL; and lenvatinib, a small-molecule TKI of VEGF and fibroblast growth factor receptors that is used in combination with everolimus, an inhibitor of the mechanistic target of rapamycin. Together, these and previously approved second-line treatments offer clinicians the ability to better individualize treatment for patients after progression on first-line VEGFR-targeted therapies. In this comprehensive review, we discuss the efficacy and safety results from the pivotal trials of these newly approved therapies, including the quality of study design, the level of evidence, subgroup analyses, and how these data can help to guide clinicians to select the most appropriate second-line therapy for their patients. IMPLICATIONS FOR PRACTICE This review article provides the reader with a comprehensive overview of current treatment options for patients with advanced clear-cell renal cell carcinoma (RCC) whose disease has progressed after their first therapy. As many patients with RCC experience disease progression with initial treatments, effective second-line therapies are critical. Nivolumab, cabozantinib, and lenvatinib plus everolimus have recently been approved as second-line treatments. The new agents discussed in this review increase the therapeutic options available and provide physicians with opportunities to individualize treatments for their patients, with a view to improving disease control and survival outcomes.
Collapse
Affiliation(s)
- Nizar M Tannir
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sumanta K Pal
- City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Michael B Atkins
- Georgetown Lombardi Comprehensive Cancer Center, Washington, D.C., USA
| |
Collapse
|
7
|
Impact of chemotherapy-induced neutropenia (CIN) and febrile neutropenia (FN) on cancer treatment outcomes: An overview about well-established and recently emerging clinical data. Crit Rev Oncol Hematol 2017; 120:163-179. [DOI: 10.1016/j.critrevonc.2017.11.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 10/20/2017] [Accepted: 11/06/2017] [Indexed: 12/13/2022] Open
|
8
|
Enzymic synthesis and biological evaluation of injectable glutathione-everolimus. Med Chem Res 2017. [DOI: 10.1007/s00044-017-2084-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
9
|
Changes in Renal Function of Patients with Metastatic Renal Cell Carcinoma During Treatment with Molecular-Targeted Agents. Target Oncol 2017; 11:329-35. [PMID: 26507837 DOI: 10.1007/s11523-015-0395-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Impairment of renal function is a serious issue that should be considered in patients undergoing treatment with molecular-targeted agents for metastatic renal cell carcinoma (mRCC). AIMS The objective of this study was to assess the impact of molecular-targeted therapy on changes in renal function among patients with mRCC. PATIENTS AND METHODS The study included 408 mRCC patients treated with sunitinib, sorafenib, axitinib, everolimus and/or temsirolimus. Among these, 185, 128 and 95 received molecular-targeted agents as first-line (group 1), second-line (group 2) and third-line (group 3) therapy, respectively. RESULTS No significant differences between the estimated glomerular filtration rate (eGFR) at baseline and that at the end of molecular-targeted therapy were noted among the three groups of patients. In addition, there were no significant differences between eGFR prior to the introduction of molecular-targeted therapy and that at the end of therapy across agents and lines of targeted therapy, with the exception of patients treated with axitinib and everolimus in second-line and third-line therapy, respectively. In group 1, a reduction in eGFR of >10 % from baseline was independently associated with performance status, hypertension and treatment duration, while in groups 2 and 3, only treatment duration was independently related to a reduction in eGFR of >10 %. CONCLUSIONS It appears that renal function in patients with mRCC is not markedly impaired by molecular-targeted therapies, irrespective of the specific agents introduced; however, it may be necessary to pay special attention to deterioration in renal function when molecular-targeted therapy is continued for longer periods.
Collapse
|
10
|
Miyake H, Imai S, Ozono S, Fujisawa M. The Inverse Association between the Baseline Renal Function and Overall Survival in Patients with Metastatic Renal Cell Carcinoma Treated with Molecular-Targeted Agents. Curr Urol 2017; 10:186-192. [PMID: 29234261 DOI: 10.1159/000447179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/15/2016] [Indexed: 01/02/2023] Open
Abstract
Background The objective of this study was to investigate the prognostic significance of the baseline renal function in metastatic renal cell carcinoma (mRCC) patients treated with molecular-targeted agents. Patients and Methods This study included 408 consecutive mRCC patients receiving molecular-targeted therapy, consisting of 124 patients in group A and 284 patients in group B who had baseline estimated glomerular filtration rates ≥ 60 ml/min/1.73 m2 and < 60 ml/min/1.73 m2, respectively. Results Compared with group A, group B was significantly less likely to have poor prognostic factors, such as a high proportion of patients without nephrectomy. The median overall survivals (OSs) after the initiation of targeted therapy in groups A and B were 21.4 and 35.8 months, respectively, and there was a significant difference in the OS between the 2 groups. However, multivariate analysis showed a lack of independent impact of the baseline renal function on the OS. Furthermore, when patients without a nephrectomy were excluded, no significant difference was noted in the OS between the 2 groups. Conclusion These findings suggested that there was no adverse impact of an unfavorable baseline renal function on the efficacy of targeted agents against mRCC. Thus, molecular-targeted therapy should not be avoided in mRCC patients with an impaired baseline renal function.
Collapse
Affiliation(s)
- Hideaki Miyake
- Department of Urology, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Satoshi Imai
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Seiichiro Ozono
- Department of Urology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masato Fujisawa
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
11
|
Abstract
Many new or relatively new cancer drugs-personalized anticancer agents-have been approved for use in various clinical settings in oncology or are still under evaluation in clinical trials. Targeted therapies as well as new immune checkpoint blockers have toxicity profiles that differ from conventional cytotoxic chemotherapy, and many can cause adverse effects that affect the mouth and pharynx, the nasal cavities, and the larynx. This review aims to provide an overview of current knowledge concerning these side effects and contemporary management. Adverse effects of the mouth/pharynx, nasal cavities, larynx, and cochlear-vestibular system are generally low grade (according to the Common Terminology Criteria for Adverse Events) and generally present non-life-threatening symptoms. However, the impact on patients' quality of life could be important. The incidence and severity vary according to the drug, its target(s), and dose, but there are currently no known predictive factors, and each patient has an individual toxicity profile. Management guidelines are based on expert opinion. These ear, nose, and throat adverse effects are not frequently mentioned in the literature because of the often non-specific nature of the symptoms and their mildness, but also the absence of specific treatment. These symptoms can contribute to decreased quality of life and lead to drug compliance issues if not diagnosed and managed appropriately.
Collapse
|
12
|
de Velasco G, Gray KP, Hamieh L, Urun Y, Carol HA, Fay AP, Signoretti S, Kwiatkowski DJ, McDermott DF, Freedman M, Pomerantz MM, Choueiri TK. Pharmacogenomic Markers of Targeted Therapy Toxicity in Patients with Metastatic Renal Cell Carcinoma. Eur Urol Focus 2016; 2:633-639. [PMID: 28723497 PMCID: PMC5520643 DOI: 10.1016/j.euf.2016.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/04/2016] [Accepted: 03/26/2016] [Indexed: 01/05/2023]
Abstract
BACKGROUND Targeted therapy (TT) in metastatic renal cell carcinoma (mRCC) may be associated with a high rate of toxicity that undermines treatment efficacy and patient quality of life. Polymorphisms in genes involved in the pharmacokinetic pathways of TTs may predict toxicity. OBJECTIVE To investigate whether selected single-nucleotide polymorphisms (SNPs) in three core genes involved in the metabolism and transport of sunitinib and the mTOR inhibitors everolimus and temsirolimus are associated with adverse events (AEs). DESIGN, SETTING, AND PARTICIPANTS Germline DNA was extracted from blood or normal kidney tissue from mRCC patients of Caucasian ethnicity in two cohorts treated with either sunitinib (n=159) or mTOR inhibitors (n=62). Six SNPs in three candidate genes (CYP3A4: rs2242480, rs4646437, and rs2246709; CYP3A5: rs15524; and ABCB1: rs2032582 and rs1045642) were analyzed. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Primary endpoints were grade ≥3 AEs for all patients; grade ≥3 hypertension in the sunitinib cohort, and any grade pneumonitis in the mTOR inhibitors cohort. A logistic regression model was used to assess the association between SNPs and AEs, with adjustment for relevant clinical factors. RESULTS AND LIMITATIONS In total, 221 samples were successfully genotyped for the selected SNPs. In the sunitinib cohort, the CYP3A4 rs464637 AG variant was associated with a lower risk of high-grade AEs (odds ratio 0.27, 95% confidence interval 0.08-0.88; p=0.03), but no SNPs were associated with hypertension. In the mTOR inhibitor cohort, none of the selected SNPs was associated with analyzed toxicities. CONCLUSIONS We observed an association between CYP3A4 polymorphisms and toxicity outcomes in mRCC patients treated with sunitinib, but not with everolimus or temsirolimus. Our findings are exploratory in nature, and further validation in independent and larger cohorts is needed. PATIENT SUMMARY We found that variants of CYP3A4, a gene involved in drug metabolism, are associated with sunitinib toxicity. This information may help in better selection of patients for targeted therapies in metastatic renal cell carcinoma.
Collapse
Affiliation(s)
| | - Kathryn P Gray
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Biostatistics and Computational Biology, Harvard School of Public Health, Boston, MA, USA
| | - Lana Hamieh
- Division of Pulmonary Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Yuksel Urun
- Department of Medical Oncology, Ankara University School of Medicine, Turkey
| | - Hallie A Carol
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Andre P Fay
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; PUCRS School of Medicine, Porto Alegre, Brazil
| | - Sabina Signoretti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - David J Kwiatkowski
- Division of Pulmonary Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - David F McDermott
- Department of Medical Oncology, Beth-Israel Deaconess Medical Center, Boston, MA, USA
| | - Matthew Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mark M Pomerantz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
13
|
Lainez N, García-Donas J, Esteban E, Puente J, Sáez MI, Gallardo E, Pinto-Marín Á, Vázquez-Estévez S, León L, García-Carbonero I, Suárez-Rodríguez C, Molins C, Climent-Duran MA, Lázaro-Quintela M, González Del Alba A, Méndez-Vidal MJ, Chirivella I, Afonso FJ, López-Brea M, Sala-González N, Domenech M, Basterretxea L, Santander-Lobera C, Gil-Arnáiz I, Fernández O, Caballero-Díaz C, Mellado B, Marrupe D, García-Sánchez J, Sánchez-Escribano R, Fernández Parra E, Villa Guzmán JC, Martínez-Ortega E, Belén González M, Morán M, Suarez-Paniagua B, Lecumberri MJ, Castellano D. Impact on clinical practice of the implementation of guidelines for the toxicity management of targeted therapies in kidney cancer. The protect-2 study. BMC Cancer 2016; 16:135. [PMID: 26906039 PMCID: PMC4763443 DOI: 10.1186/s12885-016-2084-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 01/25/2016] [Indexed: 12/21/2022] Open
Abstract
Background The impact of such recommendations after their implementation of guidelines has not usually been evaluated. Herein, we assessed the impact and compliance with the Spanish Oncology Genitourinary Group (SOGUG) Guidelines for toxicity management of targeted therapies in metastatic renal cell carcinoma (mRCC) in daily clinical practice. Methods Data on 407 mRCC patients who initiated first-line targeted therapy during the year before and the year after publication and implementation of the SOGUG guideline program were available from 34 Spanish Hospitals. Adherence to SOGUG Guidelines was assessed in every cycle. Results Adverse event (AE) management was consistent with the Guidelines as a whole for 28.7 % out of 966 post-implementation cycles compared with 23.1 % out of 892 pre-implementation cycles (p = 0.006). Analysis of adherence by AE in non-compliant cycles showed significant changes in appropriate management of hypertension (33 % pre-implementation vs. 44.5 % post-implementation cycles; p < 0.0001), diarrhea (74.0 % vs. 80.5 %; p = 0.011) and dyslipemia (25.0 % vs. 44.6 %; p < 0.001). Conclusions Slight but significant improvements in AE management were detected following the implementation of SOGUG recommendations. However, room for improvement in the management of AEs due to targeted agents still remains and could be the focus for further programs in this direction.
Collapse
Affiliation(s)
- Nuria Lainez
- Department of oncology, Complejo Hospitalario de Navarra, Servicio Oncología Médica. Pabellón B 2ª planta. Hospital de día, C/ Irunlarrea, 3, 31008, Pamplona, Navarra, Spain.
| | - Jesús García-Donas
- Department of oncology, Hospital Sanchinarro, C/ Oña, 10, 28050, Madrid, Spain.
| | - Emilio Esteban
- Department of oncology, Hospital Universitario Central de Asturias, Julián Clavería s/n, 33006, Oviedo, Spain.
| | - Javier Puente
- Department of oncology, Hospital Clínico de Madrid, C/ Doctor Martín Lagos s/n, 28040, Madrid, Spain.
| | - M Isabel Sáez
- Department of oncology, Hospital Universitario Clínico Virgen de la Victoria, Campus Universitario de Teatinos, s/n, 29010, Málaga, Spain.
| | - Enrique Gallardo
- Department of oncology, Parc Taulí Sabadell Hospital Universitari, Parc Taulí 1, 08208, Sabadell, Spain.
| | - Álvaro Pinto-Marín
- Department of oncology, Hospital Universitario La Paz, P de la Castellana 261, 28046, Madrid, Spain.
| | - Sergio Vázquez-Estévez
- Department of oncology, Hospital Universitario Lucus Augusti, Lugar San Cibrao, S/N, 27003, Lugo, Spain.
| | - Luis León
- Department of oncology, Hospital Santiago de Compostela, Travesía da Choupana, s/n, 15706, Santiago de Compostela, Spain.
| | - Icíar García-Carbonero
- Department of oncology, Hospital Virgen de la Salud, Adva. De Barber, 30, 45004, Toledo, Spain.
| | | | - Carmen Molins
- Department of oncology, Hospital Universitario Dr. Peset, Avda. Gaspar Aguilar 90, 46017, Valencia, Spain.
| | - Miguel A Climent-Duran
- Department of oncology, Instituto Valenciano de Oncología, Gregorio Gea, 31-1° Planta, 46009, Valencia, Spain.
| | - Martín Lázaro-Quintela
- Department of oncology, Complexo Hospitalario Universitario de Vigo, Pizarro 22, 36204, Vigo, Spain.
| | - Aranzazu González Del Alba
- Department of oncology, Hospital Universitario Son Espases, Ctra Valldemossa, 79, 07010, Palma de Mallorca, Spain.
| | - María José Méndez-Vidal
- Department of oncology, Hospital Universitario Reina Sofía, Avda. Menéndez Pidal, s/n, 14004, Córdoba, Spain.
| | - Isabel Chirivella
- Department of oncology, Hospital Clínico de Valencia, Avda Blasco Ibáñez, 17, 46010, Valencia, Spain.
| | - Francisco J Afonso
- Department of oncology, Complejo Hospitalario Arquitecto Marcide, Rúa da residencia s/n. San pedro de Leixa, 15405, Ferrol, Spain.
| | - Marta López-Brea
- Department of oncology, Hospital Marqués de Valdecilla, Avda. Valdecila s/n, 39008, Santander, Spain.
| | | | - Montserrat Domenech
- Department of oncology, Hospital de Althaia Xarxa Asistencial Manresa, Dr. Joan Soler, 1-3, 08243, Barcelona, Spain.
| | - Laura Basterretxea
- Department of oncology, Hospital de Donostia, P° Dr. Beguiristain 109, 20014, San Sebastian, Spain.
| | - Carmen Santander-Lobera
- Department of oncology, Hospital Miguel Servet, Avda Gómez Laguna 25, 50009, Zaragoza, Spain.
| | - Irene Gil-Arnáiz
- Department of oncology, Hospital Reina Sofía, Carretera Tarazona, KM 3, 31500, Tudela, Spain.
| | - Ovidio Fernández
- Department of oncology, Complejo Hospitalario Ourense. Hospital Santa María Nai, Ramón Puga, 52-54, 32005, Orense, Spain.
| | - Cristina Caballero-Díaz
- Department of oncology, Hospital General Universitario de Valencia, Avda Tres Cruces, s/n, 46014, Valencia, Spain.
| | - Begoña Mellado
- Department of oncology, IDIBAPS, Hospital Clinic i Provincial de Barcelona, Villarroel, 170, 08036, Barcelona, Spain.
| | - David Marrupe
- Department of oncology, Hospital Universitario de Móstoles, Río Júcar s/n, 28935, Móstoles, Madrid, Spain.
| | - José García-Sánchez
- Department of oncology, Hospital Arnau de Vilanova, C/ San Clemente n 12, 46015, Valencia, Spain.
| | - Ricardo Sánchez-Escribano
- Department of oncology, Hospital Universitario de Burgos, Avenida Cid Campeador, 96, 09005, Burgos, Spain.
| | - Eva Fernández Parra
- Department of oncology, H.U. Hospital de Valme, Ctra. de Cádiz Km. 548.9, 41014, Sevilla, Spain.
| | - José C Villa Guzmán
- Department of oncology, Hospital de Ciudad Real, Obispo Rafael Torija, 13005, Ciudad Real, Spain.
| | - Esther Martínez-Ortega
- Department of oncology, Hospital Ciudad de Jaén, Avenida Ejercito Español 10, 23007, Jaén, Spain.
| | - María Belén González
- Department of oncology, Hospital Son Llatzer, Ctra. Manacor, km.4, Sont Frriol, 07198, Palma de Mallorca, Spain.
| | - Marina Morán
- Pfizer Madrid, Avda. de Europa, 20B, 28108, Alcobendas, Madrid, Spain.
| | | | - María J Lecumberri
- Department of oncology, Complejo Hospitalario de Navarra, Servicio Oncología Médica. Pabellón B 2ª planta. Hospital de día, C/ Irunlarrea, 3, 31008, Pamplona, Navarra, Spain.
| | - Daniel Castellano
- Department of oncology, Hospital 12 de Octubre, Av. de Córdoba s/n, 28041, Madrid, Spain.
| |
Collapse
|
14
|
Patel SB, Stenehjem DD, Gill DM, Tantravahi SK, Agarwal AM, Hsu J, Vuong W, Pal SK, Agarwal N. Everolimus Versus Temsirolimus in Metastatic Renal Cell Carcinoma After Progression With Previous Systemic Therapies. Clin Genitourin Cancer 2015; 14:153-9. [PMID: 26781820 DOI: 10.1016/j.clgc.2015.12.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 12/01/2015] [Accepted: 12/09/2015] [Indexed: 12/26/2022]
Abstract
BACKGROUND Everolimus is an approved agent for use after disease progression with vascular endothelial growth factor receptor-tyrosine kinase inhibitors (VEGFR-TKIs) in patients with metastatic renal cell carcinoma. With recently published trials showing efficacy of nivolumab and cabozantinib in the second-line therapy setting, the use of everolimus will likely move to the third- or fourth-line therapy setting. Temsirolimus has occasionally been used instead of everolimus for many reasons, including financial considerations, assurance of patient compliance given its intravenous administration, its toxicity profile, patient performance status, and patient or physician preference. However, efficacy of everolimus and temsirolimus in this setting have not been compared in a randomized trial. The results from retrospective studies have been inconsistent. MATERIALS AND METHODS We identified patients treated with a first-line VEGFR-TKI for metastatic renal cell carcinoma and then treated with either everolimus or temsirolimus on progression from the databases of 2 large academic cancer centers. Progression-free survival (PFS) and overall survival (OS) were assessed from the initiation of second-line treatment using the Kaplan-Meier method. RESULTS A total of 90 patients received either everolimus (n = 59; 66%) or temsirolimus (n = 31; 34%) after progression during first-line VEGFR-TKI therapy. The patient and disease characteristics were similar in both groups. The median PFS was not different, but OS was superior with everolimus compared with temsirolimus (24.2 months vs. 12.1 months; hazard ratio, 0.58; P = .047). CONCLUSION Our results bolster existing guidelines supporting everolimus over temsirolimus as salvage therapy after previous systemic therapies.
Collapse
Affiliation(s)
- Shiven B Patel
- Department of Internal Medicine, University of Utah Huntsman Cancer Institute, Salt Lake City, UT
| | - David D Stenehjem
- Pharmacotherapy Outcomes Research Center, College of Pharmacy, The University of Utah, Salt Lake City, UT
| | - David M Gill
- Department of Internal Medicine, University of Utah Huntsman Cancer Institute, Salt Lake City, UT
| | - Srinivas K Tantravahi
- Department of Internal Medicine, University of Utah Huntsman Cancer Institute, Salt Lake City, UT
| | - Archana M Agarwal
- Department of Pathology and ARUP Laboratories, The University of Utah, Salt Lake City, UT
| | - JoAnne Hsu
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA
| | - Winston Vuong
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA
| | - Sumanta K Pal
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA
| | - Neeraj Agarwal
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT.
| |
Collapse
|
15
|
Risk factors and model for predicting toxicity-related treatment discontinuation in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted therapy: Results from the International Metastatic Renal Cell Carcin. Cancer 2015; 122:411-9. [DOI: 10.1002/cncr.29773] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 09/30/2015] [Accepted: 10/05/2015] [Indexed: 12/24/2022]
|
16
|
Miyake H, Imai S, Harada KI, Fujisawa M. Absence of Significant Correlation of Adverse Events Between First- and Second-Line Tyrosine Kinase Inhibitors in Patients With Metastatic Renal Cell Carcinoma. Clin Genitourin Cancer 2015; 14:e19-24. [PMID: 26382221 DOI: 10.1016/j.clgc.2015.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/02/2015] [Accepted: 08/08/2015] [Indexed: 12/23/2022]
Abstract
UNLABELLED Several adverse events (AEs) are known to be commonly observed during treatment with different tyrosine kinase inhibitors (TKIs) in patients with metastatic renal cell carcinoma (mRCC) patients. However, no significant correlation appears present in the profiles of such AEs between first- and second-line TKI therapies. Therefore, a second-line targeted agent for patients with mRCC could be selected irrespective of the AE profile during first-line TKI therapy. BACKGROUND Several adverse events (AEs) commonly observed during treatment with different tyrosine kinase inhibitors (TKIs). The objective of the present study was to investigate whether the appearance of such AEs during treatment with first-line TKIs significantly affects the occurrence of AEs during second-line TKI therapy for patients with metastatic renal cell carcinoma (mRCC). PATIENTS AND METHODS The present study included 154 consecutive patients with mRCC treated with second-line TKIs after the discontinuation of first-line TKIs. The association of AEs, including diarrhea, fatigue, hand-foot syndrome, hypertension, and hypothyroidism, between first- and second-line therapies was analyzed in these 154 patients. RESULTS For all 5 AEs assessed in the present study, the proportion of patients experiencing AEs or those grade ≥ 3 during second-line TKI therapy was not significantly different among the following 3 groups: patients without AEs, those with grade ≤ 2 AEs, and those with grade ≥ 3 AEs during first-line TKI therapy. Furthermore, no significant difference was seen in progression-free or overall survival after the introduction of second-line TKIs between patients with and without grade ≥ 3 AEs during treatment with first-line TKIs. CONCLUSION The incidence of AEs or grade ≥ 3 AEs during second-line TKI therapy are not dependent on the profiles of AEs during first-line TKI therapy in patients with mRCC. Therefore, AEs that occur during first-line TKI therapy should not affect the selection of second-line targeted agents for patients with mRCC.
Collapse
Affiliation(s)
- Hideaki Miyake
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Satoshi Imai
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ken-Ichi Harada
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masato Fujisawa
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
17
|
Kotecki N, Penel N, Awada A. Metabolic disorders associated with the use of targeted cancer therapies. Curr Opin Oncol 2015; 27:258-66. [PMID: 25730544 DOI: 10.1097/cco.0000000000000176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW The everyday use of targeted therapies, whose mechanisms of action differ from the conventional cytotoxic agents, also causes the emergence of new toxicities as metabolic disorders about which little is known. We propose a systematic literature review of the incidence and physiopathology of targeted therapies-induced metabolic disorders and provide some management guidance. RECENT FINDINGS In recent decades, significant breakthroughs in molecular oncology and immunology have been made. The administration of targeted therapies and immunotherapy has been associated with metabolic toxicities such as endocrine disorders, dyslipidemia, induced diabetes, and electrolytic disorders. Current data show that metabolic disorders are becoming increasingly common, but rarely life threatening and often reversible with prompt therapeutic intervention. SUMMARY In the era of targeted therapies, medical oncologists should know the symptoms, carefully monitor patients for potential metabolic disorders, and manage these emerging side-effects with the help of endocrinologists and other medical specialists.
Collapse
Affiliation(s)
- Nuria Kotecki
- aDepartment of General Oncology, Center Oscar Lambret, Lille, France bMedical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | | |
Collapse
|
18
|
Craveiro RB, Ehrhardt M, Holst MI, Pietsch T, Dilloo D. In comparative analysis of multi-kinase inhibitors for targeted medulloblastoma therapy pazopanib exhibits promising in vitro and in vivo efficacy. Oncotarget 2014; 5:7149-61. [PMID: 25216529 PMCID: PMC4196191 DOI: 10.18632/oncotarget.2240] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 07/18/2014] [Indexed: 12/12/2022] Open
Abstract
Regardless of the recent advances in cytotoxic therapies, 30% of children diagnosed with medulloblastoma. succumb to the disease. Therefore, novel therapeutic approaches are warranted. Here we demonstrate that Pazopanib a clinically approved multi-kinase angiogenesis inhibitor (MKI) inhibits proliferation and apoptosis in medulloblastoma cell lines. Moreover, Pazopanib profoundly attenuates medulloblastoma cell migration, a prerequisite for tumor invasion and metastasis. In keeping with the observed anti-neoplastic activity of Pazopanib, we also delineate reduced phosphorylation of the STAT3 protein, a key regulator of medulloblastoma proliferation and cell survival. Finally, we document profound in vivo activity of Pazopanib in an orthotopic mouse model of the most aggressive c-myc amplified human medulloblastoma variant. Pazopanib reduced the growth rate of intracranial growing medulloblastoma and significantly prolonged the survival. Furthermore, to put these results into a broader perspective we analysed Pazopanib side by side with the MKI Sorafenib. Both compounds share a similar target profile but display different pharmacodynamics and pharmacokinetics with distinct cytotoxic activity in different tumor entities. Thus, we identified Pazopanib as a new promising candidate for a rational clinical assessment for targeted paediatric medulloblastoma therapy.
Collapse
Affiliation(s)
- Rogerio B Craveiro
- Department of Pediatric Hematology and Oncology, Center for Pediatrics, University of Bonn Medical Center, Bonn, Germany. These authors contributed equally to this work
| | - Michael Ehrhardt
- Department of Pediatric Hematology and Oncology, Center for Pediatrics, University of Bonn Medical Center, Bonn, Germany. These authors contributed equally to this work
| | - Martin I Holst
- Department of Neuropathology, University of Bonn, Bonn, Germany
| | | | - Dagmar Dilloo
- Department of Pediatric Hematology and Oncology, Center for Pediatrics, University of Bonn Medical Center, Bonn, Germany
| |
Collapse
|
19
|
Strategies for the management of adverse events associated with mTOR inhibitors. Transplant Rev (Orlando) 2014; 28:126-33. [PMID: 24685370 DOI: 10.1016/j.trre.2014.03.002] [Citation(s) in RCA: 206] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 03/04/2014] [Accepted: 03/08/2014] [Indexed: 12/29/2022]
Abstract
Mammalian target of rapamycin (mTOR) inhibitors are used as potent immunosuppressive agents in solid-organ transplant recipients (everolimus and sirolimus) and as antineoplastic therapies for various cancers (eg, advanced renal cell carcinoma; everolimus, temsirolimus, ridaforolimus). Relevant literature, obtained from specific PubMed searches, was reviewed to evaluate the incidence and mechanistic features of specific adverse events (AEs) associated with mTOR inhibitor treatment, and to present strategies to effectively manage these events. The AEs examined in this review include stomatitis and other cutaneous AEs, wound-healing complications (eg, lymphocele, incisional hernia), diabetes/hyperglycemia, dyslipidemia, proteinuria, nephrotoxicity, delayed graft function, pneumonitis, anemia, hypertension, gonadal dysfunction, and ovarian toxicity. Strategies for selecting appropriate patients for mTOR inhibitor therapy and minimizing the risks of AEs are discussed, along with best practices for identifying and managing side effects. mTOR inhibitors are promising therapeutic options in immunosuppression and oncology; most AEs can be effectively detected and managed or reversed with careful monitoring and appropriate interventions.
Collapse
|
20
|
Qi H, Chen L, Liu B, Wang X, Long L, Liu D. Synthesis and biological evaluation of novel pazopanib derivatives as antitumor agents. Bioorg Med Chem Lett 2014; 24:1108-10. [DOI: 10.1016/j.bmcl.2014.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/07/2013] [Accepted: 01/04/2014] [Indexed: 01/17/2023]
|
21
|
Side-effects associated with targeted therapies in renal cell carcinoma. Curr Opin Support Palliat Care 2013; 7:254-7. [DOI: 10.1097/spc.0b013e3283644c30] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
22
|
Everolimus: side effect profile and management of toxicities in breast cancer. Breast Cancer Res Treat 2013; 140:453-62. [PMID: 23907751 DOI: 10.1007/s10549-013-2630-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 07/04/2013] [Indexed: 02/07/2023]
Abstract
Everolimus is an orally available inhibitor of the mammalian target of rapamycin (mTOR), which has been approved in combination with exemestane for hormone receptor-positive (HR) breast cancer after failure of treatment with non-steroidal aromatase inhibitors. Everolimus is generally very well tolerated with most common side effects including stomatitis, rash, fatigue, hyperglycemia, hyperlipidemia, and myelosuppression. Most of these side effects are mild and resolve with dose interruptions or dose reductions. Symptomatic non-infectious pneumonitis is a relatively uncommon class effect of mTOR inhibitors, which can be life threatening. Given the efficacy of everolimus in HR-positive metastatic breast cancer, it is crucial for physicians to recognize toxicities related to everolimus and start timely interventions. This review will focus on the adverse events reported with everolimus in breast cancer trials and will provide practical guidelines for the management of these adverse events.
Collapse
|