101
|
Gilligan T, Lin DW, Aggarwal R, Chism D, Cost N, Derweesh IH, Emamekhoo H, Feldman DR, Geynisman DM, Hancock SL, LaGrange C, Levine EG, Longo T, Lowrance W, McGregor B, Monk P, Picus J, Pierorazio P, Rais-Bahrami S, Saylor P, Sircar K, Smith DC, Tzou K, Vaena D, Vaughn D, Yamoah K, Yamzon J, Johnson-Chilla A, Keller J, Pluchino LA. Testicular Cancer, Version 2.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2020; 17:1529-1554. [PMID: 31805523 DOI: 10.6004/jnccn.2019.0058] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Testicular cancer is relatively uncommon and accounts for <1% of all male tumors. However, it is the most common solid tumor in men between the ages of 20 and 34 years, and the global incidence has been steadily rising over the past several decades. Several risk factors for testicular cancer have been identified, including personal or family history of testicular cancer and cryptorchidism. Testicular germ cell tumors (GCTs) comprise 95% of malignant tumors arising in the testes and are categorized into 2 main histologic subtypes: seminoma and nonseminoma. Although nonseminoma is the more clinically aggressive tumor subtype, 5-year survival rates exceed 70% with current treatment options, even in patients with advanced or metastatic disease. Radical inguinal orchiectomy is the primary treatment for most patients with testicular GCTs. Postorchiectomy management is dictated by stage, histology, and risk classification; treatment options for nonseminoma include surveillance, systemic therapy, and nerve-sparing retroperitoneal lymph node dissection. Although rarely occurring, prognosis for patients with brain metastases remains poor, with >50% of patients dying within 1 year of diagnosis. This selection from the NCCN Guidelines for Testicular Cancer focuses on recommendations for the management of adult patients with nonseminomatous GCTs.
Collapse
Affiliation(s)
- Timothy Gilligan
- 1Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | - Daniel W Lin
- 2University of Washington/Seattle Cancer Care Alliance
| | | | | | | | | | | | | | | | | | | | | | | | - Will Lowrance
- 14Huntsman Cancer Institute at the University of Utah
| | | | - Paul Monk
- 16The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Joel Picus
- 17Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | | | | | | | - Daniel Vaena
- 24St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | - David Vaughn
- 25Abramson Cancer Center at the University of Pennsylvania
| | | | | | | | | | | |
Collapse
|
102
|
Cardoso AR, Lobo J, Miranda-Gonçalves V, Henrique R, Jerónimo C. Epigenetic alterations as therapeutic targets in Testicular Germ Cell Tumours : current and future application of 'epidrugs'. Epigenetics 2020; 16:353-372. [PMID: 32749176 DOI: 10.1080/15592294.2020.1805682] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Testicular germ cell tumours (TGCTs) are heterogeneous neoplasms mostly affecting young-adult men. Despite high survival rates, some patients with disseminated disease acquire cisplatin resistance, entailing the need for less toxic therapies. Epigenetic alterations constitute an important feature of TGCTs, which are also implicated in resistance mechanism(s). These alterations might be used as potential targets to design epigenetic drugs. To date, several compounds have been explored and evaluated regarding therapeutic efficacy, making use of pre-clinical studies with in vitro and in vivo models, and some have already been explored in clinical trials. This review summarizes the several epigenetic mechanisms at play in these neoplasms, the current challenges in the field of TGCTs and critically reviews available data on 'epidrugs' in those tumours.
Collapse
Affiliation(s)
- Ana Rita Cardoso
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), 4200-072, Porto, Portugal.,Master in Oncology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), 4050-513, Porto, Portugal
| | - João Lobo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), 4200-072, Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), 4200-072, Porto, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), 4050-513, Porto, Portugal
| | - Vera Miranda-Gonçalves
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), 4200-072, Porto, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), 4050-513, Porto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), 4200-072, Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), 4200-072, Porto, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), 4050-513, Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), 4200-072, Porto, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), 4050-513, Porto, Portugal
| |
Collapse
|
103
|
Callec L, Lardy-Cleaud A, Guerrini-Rousseau L, Alapetite C, Vignon L, Chastagner P, Frappaz D, Faure-Conter C. Relapsing intracranial germ cell tumours warrant retreatment. Eur J Cancer 2020; 136:186-194. [PMID: 32711377 DOI: 10.1016/j.ejca.2020.06.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 05/19/2020] [Accepted: 06/08/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND The optimal therapeutic strategy for relapsing intracranial germ cell tumours (IGCTs) has not been clearly established. METHODS Relapses of IGCTs, occurring from 01/01/1990 to 31/12/2014, were retrieved from the Societe Française d'Oncologie Pediatrique-TGM 90, 92 and GCT 96 protocols, and from the National Childhood Solid Tumour Registry. Refractory IGCTs were excluded. RESULTS Forty-four relapsing IGCTs were identified: 14 were initially treated for histologically proven germinomas (germinoma group), 5 for non-histologically proven germinomas (putative germinoma group) and 25 for non-germinomatous germ cell tumours (NGGCTs) (NGGCT group). In the germinoma group, the 5-year event-free survival (EFS) and overall survival (OS) were 79% (95% confidence interval [CI]: 47-93) and 86% (95% CI: 54-96), respectively. Only one of the 11 patients treated with reirradiation experienced a further relapse. A trend of better EFS was observed for relapses at sites that were not initially involved: 5-year EFS of 100% versus 67% (95% CI: 28-88), p = 0.09. In the putative germinoma group, 4 of 5 patients experienced a further event, leading to 2 deaths. In the NGGCT group, the 5-year EFS and OS were 56% (95% CI: 35-73) and 60% (95% CI: 38-76), respectively. A significant improvement in outcomes after high-dose chemotherapy (HDC) was observed: 5-year OS of 72% (95% CI: 46-87) versus 29% (95% CI: 4-61), p = 0.006. CONCLUSION Relapsing germinomas are highly curable; reirradiation appears to play a key role. Histological proof at initial diagnosis if markers are negative is crucial. Despite inferior outcomes relapsing, NGGCTs can be cured in a significant proportion of cases provided intensive treatment including HDC is applied.
Collapse
Affiliation(s)
- Laetitia Callec
- Laetitia Callec, CHU- Hôpitaux de Brabois, Department of Pediatrics, Vandoeuvre les Nancy, France
| | - Audrey Lardy-Cleaud
- Audrey Lardy-Cleaud, Department of Biostatistic, Centre Léon Bérard, Lyon, France
| | - Lea Guerrini-Rousseau
- Lea Guerrini-Rousseau, Department of Pediatric and Adolescents Oncology, Institut Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - Claire Alapetite
- Claire Alapetite, Institut Curie, Department of Radiation Oncology, Paris, France
| | - Laure Vignon
- Lucie Vignon, INSERM, Paris-Descartes University, Department of Epidemiology, French National Registry of Childhood Solid Tumour Registry (RNTSE), Villejuif, France
| | - Pascal Chastagner
- Pascal Chastagner, CHU- Hôpitaux de Brabois, Department of Pediatrics, Vandoeuvre les Nancy, France
| | - Didier Frappaz
- Didier Frappaz, Institut d'Hemato-oncologie Pediatrique, Lyon, France
| | | |
Collapse
|
104
|
Molecular Pathways and Targeted Therapies for Malignant Ovarian Germ Cell Tumors and Sex Cord-Stromal Tumors: A Contemporary Review. Cancers (Basel) 2020; 12:cancers12061398. [PMID: 32485873 PMCID: PMC7353025 DOI: 10.3390/cancers12061398] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/13/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022] Open
Abstract
Non-epithelial ovarian tumors are heterogeneous and account for approximately 10% of ovarian malignancies. The most common subtypes of non-epithelial ovarian tumors arise from germ cells or sex cord and stromal cells of the gonads. These tumors are usually detected at an early stage, and management includes surgical staging and debulking. When indicated for advanced disease, most respond to chemotherapy; however, options for patients with refractory disease are limited, and regimens can be associated with significant toxicities, including permanent organ dysfunction, secondary malignancies, and death. Targeted therapies that potentially decrease chemotherapy-related adverse effects and improve outcomes for patients with chemotherapy-refractory disease are needed. Here, we review the molecular landscape of non-epithelial ovarian tumors for the purpose of informing rational clinical trial design. Recent genomic discoveries have uncovered recurring somatic alterations and germline mutations in subtypes of non-epithelial ovarian tumors. Though there is a paucity of efficacy data on targeted therapies, such as kinase inhibitors, antibody–drug conjugates, immunotherapy, and hormonal therapy, exceptional responses to some compounds have been reported. The rarity and complexity of non-epithelial ovarian tumors warrant collaboration and efficient clinical trial design, including high-quality molecular characterization, to guide future efforts.
Collapse
|
105
|
Spears N, Lopes F, Stefansdottir A, Rossi V, De Felici M, Anderson RA, Klinger FG. Ovarian damage from chemotherapy and current approaches to its protection. Hum Reprod Update 2020; 25:673-693. [PMID: 31600388 PMCID: PMC6847836 DOI: 10.1093/humupd/dmz027] [Citation(s) in RCA: 383] [Impact Index Per Article: 76.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/18/2019] [Accepted: 07/23/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Anti-cancer therapy is often a cause of premature ovarian insufficiency and infertility since the ovarian follicle reserve is extremely sensitive to the effects of chemotherapy and radiotherapy. While oocyte, embryo and ovarian cortex cryopreservation can help some women with cancer-induced infertility achieve pregnancy, the development of effective methods to protect ovarian function during chemotherapy would be a significant advantage. OBJECTIVE AND RATIONALE This paper critically discusses the different damaging effects of the most common chemotherapeutic compounds on the ovary, in particular, the ovarian follicles and the molecular pathways that lead to that damage. The mechanisms through which fertility-protective agents might prevent chemotherapy drug-induced follicle loss are then reviewed. SEARCH METHODS Articles published in English were searched on PubMed up to March 2019 using the following terms: ovary, fertility preservation, chemotherapy, follicle death, adjuvant therapy, cyclophosphamide, cisplatin, doxorubicin. Inclusion and exclusion criteria were applied to the analysis of the protective agents. OUTCOMES Recent studies reveal how chemotherapeutic drugs can affect the different cellular components of the ovary, causing rapid depletion of the ovarian follicular reserve. The three most commonly used drugs, cyclophosphamide, cisplatin and doxorubicin, cause premature ovarian insufficiency by inducing death and/or accelerated activation of primordial follicles and increased atresia of growing follicles. They also cause an increase in damage to blood vessels and the stromal compartment and increment inflammation. In the past 20 years, many compounds have been investigated as potential protective agents to counteract these adverse effects. The interactions of recently described fertility-protective agents with these damage pathways are discussed. WIDER IMPLICATIONS Understanding the mechanisms underlying the action of chemotherapy compounds on the various components of the ovary is essential for the development of efficient and targeted pharmacological therapies that could protect and prolong female fertility. While there are increasing preclinical investigations of potential fertility preserving adjuvants, there remains a lack of approaches that are being developed and tested clinically.
Collapse
Affiliation(s)
- N Spears
- Biomedical Sciences, University of Edinburgh, Edinburgh UK
| | - F Lopes
- Biomedical Sciences, University of Edinburgh, Edinburgh UK
| | | | - V Rossi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - M De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - R A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh UK
| | - F G Klinger
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
106
|
Ray S, Pierorazio PM, Allaf ME. Primary and post-chemotherapy robotic retroperitoneal lymph node dissection for testicular cancer: a review. Transl Androl Urol 2020; 9:949-958. [PMID: 32420211 PMCID: PMC7214990 DOI: 10.21037/tau.2020.02.09] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Open retroperitoneal lymph node dissection (RPLND) is the gold standard for surgical management of the retroperitoneum in patients with testicular cancer, and is associated with excellent oncologic outcomes and significant morbidity including length of stay. Minimally invasive RPLND, starting with laparoscopic retroperitoneal lymph node dissection in 1992 and now robotic retroperitoneal lymph node dissection in 2006, endeavor to decrease the morbidity of open RPLND while maintaining excellent oncologic outcomes. This review surveys the literature regarding both primary and post-chemotherapy robotic RPLND, emphasizing that while early outcomes are promising, much work needs to be done before widespread use of this technique is implemented.
Collapse
Affiliation(s)
- Shagnik Ray
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Phillip M Pierorazio
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mohamad E Allaf
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
107
|
Labbate CV, Werntz RP, Galansky LB, Packiam VT, Eggener SE. National management trends in clinical stage IIA nonseminomatous germ cell tumor (NSGCT) and opportunities to avoid dual therapy. Urol Oncol 2020; 38:687.e13-687.e18. [PMID: 32305267 DOI: 10.1016/j.urolonc.2020.03.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/08/2020] [Accepted: 03/17/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION For marker-negative clinical stage (CS) IIA nonseminomatous germ cell tumor (NSGCT), National Comprehensive Cancer Network and American Urological Association guidelines recommend either retroperitoneal lymph node dissection (RPLND) or induction chemotherapy. The goal is cure with one form of therapy. We evaluated national practice patterns in the management of CSIIA NSGCT and utilization of secondary therapies. METHODS The National Cancer Data Base was used to identify 400 men diagnosed with marker negative CSIIA NSGCT between 2004 and 2014 treated with RPLND or chemotherapy. Trends in the utilization of initial and adjuvant treatment (chemotherapy only, RPLND only, RPLND with adjuvant chemotherapy, and postchemotherapy RPLND) were analyzed. RESULTS Of the 400 cases, 233 (58%) underwent induction chemotherapy with surveillance, 51 (20%) underwent RPLND with surveillance, 89 (22%) underwent RPLND followed by adjuvant chemotherapy, and 14 (4%) underwent induction chemotherapy followed by RPLND. Thirty percent of patients received dual therapy. After RPLND with pN1 staging, 43 (61%) underwent adjuvant chemotherapy. The pN0 rate after primary RPLND was 22%. Five year overall survival ranged from 95% to 100% based on initial treatment choice. CONCLUSIONS For marker negative CS IIA nonseminoma, dual, therapy, and treatment with chemotherapy is common. With low volume retroperitoneal disease resected at RPLND, adjuvant chemotherapy was frequently administered but has debatable therapeutic value. These data highlight opportunities to decrease treatment burden in patients with CS IIA nonseminoma.
Collapse
Affiliation(s)
- Craig V Labbate
- Section of Urology, University of Chicago Medicine, Chicago, IL.
| | - Ryan P Werntz
- Prisma Health-Upstate, Section of Urology, Department of Surgery, University of South Carolina-Greenville
| | | | | | - Scott E Eggener
- Section of Urology, University of Chicago Medicine, Chicago, IL
| |
Collapse
|
108
|
Furubayashi N, Negishi T, Takamatsu D, Ieiri K, Inoue T, Tsukino K, Nakamura M. Timing of changing therapy from gemcitabine and cisplatin chemotherapy based on real-world data of advanced urothelial carcinoma. Oncol Lett 2020; 19:2943-2949. [PMID: 32256805 PMCID: PMC7074169 DOI: 10.3892/ol.2020.11368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 01/21/2020] [Indexed: 12/17/2022] Open
Abstract
Cisplatin-based systemic chemotherapy is the gold-standard approach for the first-line treatment of patients with advanced or metastatic urothelial carcinoma (UC). However, the optimal number of cycles is still unclear. The current study retrospectively assessed the clinical outcome in patients who received gemcitabine and cisplatin (GC) chemotherapy as first-line treatment for metastatic urothelial cancer to clarify the timing of switching from GC therapy. A total of 61 patients with locally advanced or metastatic UC who received first-line chemotherapy with GC were retrospectively reviewed at National Hospital Organization Kyushu Cancer Center between June 2009 and August 2017. The progression-free survival (PFS) and overall survival (OS) were evaluated using the Kaplan-Meier method. The significance of associations between the clinical parameters and OS was assessed using the Cox proportional hazards regression model. The median cycle number for GC chemotherapy was 4. The median PFS and OS of all cases was 5.2 and 14.1 months, respectively. The multivariate analyses revealed that a neutrophil-to-lymphocyte ratio ≥3.0 (hazard ratio [HR], 2.521, 95% confidence interval [CI]=1.179-5.624; P=0.017) and best response to GC therapy of CR+PR (HR 0.110; 95% CI=0.028-0.411; P<0.001) were independent prognostic factors. However, the number of GC cycles (≤4 vs. >4) was not an independent prognostic factor (P=0.387). The current retrospective study indicated that changes to therapy should be considered at an early stage for cases with a therapeutic effect of SD or less, regardless of the number of GC therapy cycles.
Collapse
Affiliation(s)
- Nobuki Furubayashi
- Department of Urology, National Hospital Organization Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Takahito Negishi
- Department of Urology, National Hospital Organization Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Dai Takamatsu
- Department of Urology, National Hospital Organization Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Kosuke Ieiri
- Department of Urology, National Hospital Organization Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Tomohiro Inoue
- Department of Urology, National Hospital Organization Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Keiji Tsukino
- Department of Urology, National Hospital Organization Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Motonobu Nakamura
- Department of Urology, National Hospital Organization Kyushu Cancer Center, Fukuoka 811-1395, Japan
| |
Collapse
|
109
|
Rabaça A, Ferreira C, Bernardino R, Alves M, Oliveira P, Viana P, Barros A, Sousa M, Sá R. Use of antioxidant could ameliorate the negative impact of etoposide on human sperm DNA during chemotherapy. Reprod Biomed Online 2020; 40:856-866. [PMID: 32376314 DOI: 10.1016/j.rbmo.2020.01.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 12/31/2019] [Accepted: 01/30/2020] [Indexed: 01/07/2023]
Abstract
RESEARCH QUESTION A previous study showed that N-acetylcysteine (NAC), used after in-vitro exposure to the gonadotoxic chemotherapeutic drug etoposide, has the ability to decrease DNA damage in human spermatozoa; however, it showed no benefit when used before exposure. This study aimed to evaluate the impact of the NAC on the preservation of sperm quality during in-vitro exposure to etoposide. DESIGN Twenty semen samples were submitted to four experimental conditions: control, NAC-only incubation, etoposide-only incubation, and concomitant etoposide and NAC incubation. After in-vitro incubation, semen parameters, sperm chromatin condensation, sperm DNA fragmentation, sperm oxidative stress and sperm metabolism were used to evaluate the role of NAC in protecting human spermatozoa from etoposide. RESULTS Etoposide did not affect semen parameters, nor did it cause sperm oxidative damage or alterations in glycolytic profile. However, it induced chromatin decondensation and DNA fragmentation, which were fully prevented by NAC. CONCLUSIONS NAC was able to protect sperm DNA integrity during etoposide treatment in vitro, suggesting that NAC may be useful as an adjuvant agent in preserving male fertility during chemotherapy treatments.
Collapse
Affiliation(s)
- Ana Rabaça
- Laboratory of Cell Biology, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (UP), Porto, Portugal
| | - Carolina Ferreira
- Laboratory of Cell Biology, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (UP), Porto, Portugal
| | - Raquel Bernardino
- Laboratory of Cell Biology, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (UP), Porto, Portugal; Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
| | - Marco Alves
- Laboratory of Cell Biology, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (UP), Porto, Portugal; Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
| | - Pedro Oliveira
- Laboratory of Cell Biology, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (UP), Porto, Portugal; Department of Genetics, Faculty of Medicine, University of Porto, Porto, Portugal; Health Institute of Research and Innovation (IPATIMUP/i3S), University of Porto, Porto, Portugal
| | - Paulo Viana
- Centre for Reproductive Genetics A. Barros (CGR), Porto, Portugal
| | - Alberto Barros
- Department of Genetics, Faculty of Medicine, University of Porto, Porto, Portugal; Health Institute of Research and Innovation (IPATIMUP/i3S), University of Porto, Porto, Portugal; Centre for Reproductive Genetics A. Barros (CGR), Porto, Portugal
| | - Mário Sousa
- Laboratory of Cell Biology, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (UP), Porto, Portugal; Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal.
| | - Rosália Sá
- Laboratory of Cell Biology, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (UP), Porto, Portugal; Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
| |
Collapse
|
110
|
Qin Z, Ren G, Yuan J, Chen H, Lu Y, Li N, Zhang Y, Chen X, Zhao D. Systemic Evaluation on the Pharmacokinetics of Platinum-Based Anticancer Drugs From Animal to Cell Level: Based on Total Platinum and Intact Drugs. Front Pharmacol 2020; 10:1485. [PMID: 31969818 PMCID: PMC6960190 DOI: 10.3389/fphar.2019.01485] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/15/2019] [Indexed: 12/20/2022] Open
Abstract
Cisplatin, carboplatin, and oxaliplatin are the common platinum-based anticancer drugs widely used in the chemotherapeutic treatment of solid tumors in clinic. However, the comprehensive pharmacokinetics of platinum-based anticancer drugs has not been fully understood yet. This leads to many limitations for the further studies on their pharmacology and toxicology. In this study, we conduct a systemic evaluation on the pharmacokinetics of three platinum analogues at animal and cell levels, with quantification of both total platinum and intact drugs. A detailed animal study to address and compare the different pharmacokinetic behaviors of three platinum analogues has been conducted in three biological matrices: blood, plasma, and ultrafiltrate plasma. Carboplatin showed an obviously different pharmacokinetic characteristic from cisplatin and oxaliplatin. On the one hand, carboplatin has the highest proportion of Pt distribution in ultrafiltrate plasma. On the other hand, carboplatin has the highest intact drug exposure and longest intact drug elimination time in blood, plasma, and ultrafiltrate plasma, which may explain its high hematotoxicity. Additionally, the cellular and subcellular pharmacokinetics of oxaliplatin in two colon cancer HCT-116/LOVO cell lines has been elucidated for the first time. The biotransformation of intact oxaliplatin in cells was rapid with a fast elimination, however, the generated platinum-containing metabolites still exist within cells. The distribution of total platinum in the cytosol is higher than in the mitochondria, followed by the nucleus. Enrichment of platinum in mitochondria may affect the respiratory chain or energy metabolism, and further lead to cell apoptosis, which may indicate mitochondria as another potential target for efficacy and toxicity of oxaliplatin.
Collapse
Affiliation(s)
- Zhiying Qin
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Guanghui Ren
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Jinjie Yuan
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Huili Chen
- School of Engineering & Applied Science, Yale University, New Haven, CT, United States
| | - Yang Lu
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Ning Li
- National Experimental Teaching Demonstration Center of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yongjie Zhang
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Xijing Chen
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Di Zhao
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
111
|
Zhang Y, Zheng J, Jiang Y, Huang X, Fang L. Neglected, Drug-Induced Platinum Accumulation Causes Immune Toxicity. Front Pharmacol 2020; 11:1166. [PMID: 32903504 PMCID: PMC7438596 DOI: 10.3389/fphar.2020.01166] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/17/2020] [Indexed: 02/05/2023] Open
Abstract
Previous studies only focused on different adverse reactions caused by various platinum drugs, but not on common immunotoxicity caused by the accumulation of elemental platinum. Here, we determined the serum platinum concentrations of cancer patients after a metabolism period of platinum drug chemotherapy, in addition to hematological indices and subsequent immune-related adverse reactions, then analyzed the correlations between platinum accumulation, immune cell levels, and immune-toxicity. We chose the day before the next round of chemotherapy as the specified time point for blood sampling. Samples were collected at five time points, separately in oxaliplatin and cisplatin groups. The median serum platinum concentrations in all patients was 294.8 (205.6, 440.3) μg/L, and was approximately two-fold greater in the cisplatin group than in the oxaliplatin group (429.3 vs. 211.7 μg/L). The platinum level of both groups peaked at the third time point, with the average of females being higher than males (383.9 vs. 266.5 μg/L), and was positively correlated with leukocyte and platelet counts, but negatively correlated with erythrocyte counts and concentration of hemoglobin. The risks of anemia and adverse reactions were individually increased by 0.002- and 0.007-fold for every μg/L increase of platinum concentration. To our knowledge, this is the first study on the relationship between platinum accumulation, immune cell levels and toxicity, showing that drug-induced platinum accumulation may interfere with immune cells and thus increase the risk of toxicity.
Collapse
Affiliation(s)
- Yuling Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, China
| | - Jieting Zheng
- Pharmacy Department, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yi Jiang
- Digestive Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Xuchun Huang
- Department of Clinical Laboratory Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Ling Fang
- Pharmacy Intravenous Admixture Service, Cancer Hospital of Shantou University Medical College, Shantou, China
- *Correspondence: Ling Fang,
| |
Collapse
|
112
|
Prabhash K, Patil V, Noronha V, Joshi A, Talreja V, Dhumal S, Menon N, Abhyankar A, Dsouza H, Singh G, Bhattacharjee A, Ghosh-Laskar S, Pai P, Chaturvedi P, Nair D, Chaukar D, DCruz A, Shetty P, Moiyadi A. Long-term outcomes of locally advanced and borderline resectable esthesioneuroblastoma and sinonasal tumor with neuroendocrine differentiation treated with neoadjuvant chemotherapy. CANCER RESEARCH, STATISTICS, AND TREATMENT 2020. [DOI: 10.4103/crst.crst_78_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
113
|
Kvammen Ø, Myklebust TÅ, Solberg A, Møller B, Klepp OH, Fosså SD, Tandstad T. Causes of inferior relative survival after testicular germ cell tumor diagnosed 1953-2015: A population-based prospective cohort study. PLoS One 2019; 14:e0225942. [PMID: 31851716 PMCID: PMC6919610 DOI: 10.1371/journal.pone.0225942] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 11/15/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Testicular germ cell tumor (TGCT) patients and survivors have excess mortality compared to the general male population, but relative survival (RS) has been scarcely studied. We investigated causes of excess mortality and their impact on RS among men diagnosed with TGCT in Norway, 1953-2015. METHODS AND FINDINGS Using registry data (n = 9541), standardized mortality ratios (SMRs) and RS were calculated. By December 31st, 2015, 816 testicular cancer (TC) and 1508 non-TC deaths had occurred (non-TC SMR: 1.36). Within five years of TGCT diagnosis, 80% were TC deaths. Non-TC second cancer (SC) caused 65% of excess non-TC deaths, of which 34% from gastric, pancreatic or bladder cancer. SC SMRs remained elevated ≥26 years of follow-up. In localized TGCT diagnosed >1979, SC SMRs were only elevated after seminoma. Cardiovascular disease caused 9% and other causes 26% of excess non-TC deaths, of which 58% from gastrointestinal and genitourinary disorders. RS continuously declined with follow-up. TGCT patients diagnosed >1989 had superior five-year TC-specific RS (98.3%), lower non-TC SMR (1.21), but elevated SMRs for several SCs, infections, Alzheimer's disease, genitourinary disease and suicide. A limitation was lack of individual treatment data. CONCLUSIONS RS declines mainly from TC deaths <5 years after TGCT diagnosis. Later, excess SC mortality becomes particularly important, reducing RS even ≥26 years. Radiotherapy; standard adjuvant seminoma treatment 1980-2007, is likely an important contributor, as are chemotherapy and possibly innate susceptibilities. Vigilant long-term follow-up, including psychosocial aspects, is important. Further research should focus on identifying survivor risk groups and optimizing treatment.
Collapse
Affiliation(s)
- Øivind Kvammen
- Department of Oncology, Ålesund Hospital, Ålesund, Norway
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Tor Åge Myklebust
- Department of Research and Innovation, Møre and Romsdal Hospital Trust, Ålesund, Norway
- Department of Registration, Cancer Registry of Norway, Oslo, Norway
| | - Arne Solberg
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- The Cancer Clinic, St. Olav´s University Hospital, Trondheim, Norway
| | - Bjørn Møller
- Department of Registration, Cancer Registry of Norway, Oslo, Norway
| | | | - Sophie Dorothea Fosså
- National Advisory Unit on Late Effects after Cancer Treatment, Oslo University Hospital, The Radium Hospital, Oslo, Norway
- Faculty of Medicine, Oslo University, Oslo, Norway
| | - Torgrim Tandstad
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- The Cancer Clinic, St. Olav´s University Hospital, Trondheim, Norway
| |
Collapse
|
114
|
Lopes F, Liu J, Morgan S, Matthews R, Nevin L, Anderson RA, Spears N. Single and combined effects of cisplatin and doxorubicin on the human and mouse ovary in vitro. Reproduction 2019; 159:193-204. [PMID: 31821159 PMCID: PMC6993208 DOI: 10.1530/rep-19-0279] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 12/09/2019] [Indexed: 01/06/2023]
Abstract
Chemotherapy drugs are administered to patients using combination regimens, and as such the possibility of multiplicative effects between drugs need to be investigated. This study examines the individual and combined effects of the chemotherapy drugs cisplatin and doxorubicin on the human ovary. Although cisplatin and doxorubicin are known to affect female fertility, there is limited information about their direct effects on the human ovary, and none examining the possibility of combined, multiplicative effects of co-exposure to these drugs. Here, human ovarian biopsies were obtained from 14 women at the time of caesarean section, with 38 mouse ovaries also obtained from neonatal C57Bl/6J mice. Tissue was cultured for 6 days prior to analyses, with chemotherapy drugs added to culture medium on the second day of culture only. Treatment groups of a single (5 μg/mL human; 0.5 μg/mL mouse) or double (10 μg/mL human; 1.0 μg/mL mouse) dose of cisplatin, a single (1 μg/mL human; 0.05 μg/mL mouse) or double (2 μg/mL human; 0.1 μg/mL mouse) dose of doxorubicin or a combination of a single dose of both drugs together were compared to controls without drug exposure. Exposure to cisplatin or doxorubicin significantly decreased follicle health in human and mouse, supporting the suitability of the mouse as a model for the human ovary. There was also a significant reduction of mouse follicle number. Human ovarian stromal tissue exhibited increased apoptosis and decreased cell proliferation. Crucially, there was no evidence indicating the occurrence of multiplicative effects between cisplatin and doxorubicin.
Collapse
Affiliation(s)
- Federica Lopes
- F Lopes, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom of Great Britain and Northern Ireland
| | - Jin Liu
- J Liu, Department of Public Health, Fujian Medical University, Fuzhou, China
| | - Stephanie Morgan
- S Morgan, Biological Sciences, University of Southampton, Southampton, United Kingdom of Great Britain and Northern Ireland
| | - Rebecca Matthews
- R Matthews, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom of Great Britain and Northern Ireland
| | - Lucy Nevin
- L Nevin, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom of Great Britain and Northern Ireland
| | - Richard A Anderson
- R Anderson, MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom of Great Britain and Northern Ireland
| | - Norah Spears
- N Spears, Biomedical Sciences, University of Edinburgh, Edinburgh, EH8 9XD, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
115
|
Influence of steric and electronic effect of carrier ligand on kinetics & mechanism of Pt(II) complexes with l-cysteine and its substituted derivatives: Their experimental and DFT-based theoretical study. Inorganica Chim Acta 2019. [DOI: 10.1016/j.ica.2019.119117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
116
|
Kreiberg M, Bandak M, Lauritsen J, Andersen KK, Skøtt JW, Johansen C, Agerbaek M, Holm NV, Lau CJ, Daugaard G. Psychological stress in long-term testicular cancer survivors: a Danish nationwide cohort study. J Cancer Surviv 2019; 14:72-79. [PMID: 31748852 DOI: 10.1007/s11764-019-00835-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/12/2019] [Indexed: 01/10/2023]
Abstract
PURPOSE Long-term cancer survivors may develop psychological late effects. The aim of the present study was to determine prevalence of high level of stress in testicular cancer survivors (TCS) compared with the general population and prevalence of high level of stress among TCS stratified by type of treatment (surveillance, bleomycin, etoposide and cisplatin (BEP), or abdominal radiotherapy (RT)). METHODS In this large, nationwide and population-based, cross-sectional study, a total of 2252 TCS filled in a questionnaire between 2014-2016 covering psychological stress (Perceived Stress Scale (PSS)), sociodemographic factors, and physical health variables. Results were compared with a reference population. The reference population consisted of 61,927 men without prior or present cancer and sampled at random from the central population. High level of stress was defined as a PSS score ≥ 16, equivalent to the highest scoring quintile in the reference population. Logistic regression models adjusted for relevant covariates were used to estimate prevalence ratios of high level of stress. RESULTS Distribution of TCS was: surveillance, n = 1134; BEP, n = 807; and RT, n = 311 (median time since diagnosis was 19 years). TCS were more likely to have high level of stress compared to the reference population with a prevalence ratio of 1.56 (95% CI, 1.40-1.73). Individually, surveillance, BEP and RT groups had higher level of stress compared to the reference population. CONCLUSIONS TCS are more likely to have high level of stress. Screening programs for psychological stress should be considered as part of the follow-up program. IMPLICATIONS FOR CANCER SURVIVORS A higher level of stress is observed in TCS irrespective of treatment.
Collapse
Affiliation(s)
- Michael Kreiberg
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, 2100, Copenhagen, Denmark.
| | - Mikkel Bandak
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, 2100, Copenhagen, Denmark
| | - Jakob Lauritsen
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, 2100, Copenhagen, Denmark
| | - Klaus Kaae Andersen
- Department of Statistics and Pharmacoepidemiology, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Julie Wang Skøtt
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, 2100, Copenhagen, Denmark
| | - Christoffer Johansen
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, 2100, Copenhagen, Denmark.,Unit of Survivorship, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Mads Agerbaek
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Niels V Holm
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Cathrine Juel Lau
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, The Capital Region, Copenhagen, Denmark
| | - Gedske Daugaard
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, 2100, Copenhagen, Denmark
| |
Collapse
|
117
|
Chovanec M, Kalavska K, Mego M, Cheng L. Liquid biopsy in germ cell tumors: biology and clinical management. Expert Rev Mol Diagn 2019; 20:187-194. [PMID: 31652083 DOI: 10.1080/14737159.2019.1685383] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction: Liquid biopsy is an increasingly studied approach for optimal and minimally invasive diagnostics of malignant tumors. The aim of this review is to provide evidence and discuss the utility of liquid biopsy in the management of germ cell tumors (GCTs).Areas covered: Herein, we summarize the evidence on liquid biopsy in GCTs including serum tumor markers, circulating tumor cells, microRNA and cell-free DNA. The search of literature was conducted from Pubmed/Medline, ASCO-meeting library searching for terms 'liquid biopsy', 'germ cell tumors', 'circulating tumor cells', 'microRNA', 'cell-free DNA'. Obtained original studies were included. Reference lists of review articles and key original articles were searched for additional original studies. We included articles published between1990 and 2019.Expert opinion: Liquid biopsy is a minimally invasive tool using body fluids for diagnostic purposes in cancer. The established value of serum tumor markers may be already considered a liquid biopsy technique in diagnosis of GCTs. Possible near-future refinements in diagnosis of GCTs are emerging. Further information on diagnosis, prognosis and resistance is added with recently described microRNAs, circulating tumor cells and cell-free DNA. While great promise is shown, further large-scale validation is needed to incorporate these novel liquid biopsies into clinical practice.
Collapse
Affiliation(s)
- Michal Chovanec
- 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia.,Division of Hematology Oncology, Indiana University Simon Cancer Center, Indianapolis, IN, USA
| | - Katarina Kalavska
- Translational Research Unit at 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Michal Mego
- 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia.,Translational Research Unit at 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine0, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Urology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
118
|
New Insights in the Pathogenesis of Cisplatin-Induced Nephrotoxicity. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2019. [DOI: 10.2478/sjecr-2019-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Cisplatin (cis-diamminedichloroplatinum II) is a widely used chemotherapeutic agent. However, efficacy and clinical utility of this drug is significantly limited by severe side effects such as nephrotoxicity which develops due to renal accumulation and bio-transformation in proximal tubular epithelial cells. Cisplatin-induced nephrotoxicity can be manifested as acute kidney injury (AKI), or as different types of tubulopathies, salt wasting, loss of urinary concentrating ability, and magnesium wasting. The attenuation of cisplatin-caused AKI is currently accomplished by hydration, magnesium supplementation or mannitol-induced forced diuresis. However, mannitol treatment causes over-diuresis and consequent dehydration, indicating an urgent need for the clinical use of newly designed, safe and efficacious renoprotective drug, as an additive therapy for high dose cisplatin-treated patients. Accordingly, we emphasized current knowledge regarding molecular mechanisms responsible for cisplatin-caused nephrotoxicity and we described in detail the main clinical manifestations of cisplatin-induced renal dysfunction in order to pave the way for the design of new therapeutic approaches that can minimize detrimental effects of cisplatin in the kidneys. Having in mind that most of cisplatin-induced cytotoxic effects against renal cells are, at the same time, involved in anti-tumor activity of cisplatin, new nephroprotective therapeutic strategies have to prevent renal injury and inflammation without affecting cisplatin-induced toxicity against malignant cells.
Collapse
|
119
|
Trendowski MR, El-Charif O, Ratain MJ, Monahan P, Mu Z, Wheeler HE, Dinh PC, Feldman DR, Ardeshir-Rouhani-Fard S, Hamilton RJ, Vaughn DJ, Fung C, Kollmannsberger C, Mushiroda T, Kubo M, Hannigan R, Strathmann F, Einhorn LH, Fossa SD, Travis LB, Dolan ME. Clinical and Genome-Wide Analysis of Serum Platinum Levels after Cisplatin-Based Chemotherapy. Clin Cancer Res 2019; 25:5913-5924. [PMID: 31296530 PMCID: PMC6774840 DOI: 10.1158/1078-0432.ccr-19-0113] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/17/2019] [Accepted: 07/09/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE Serum platinum is measurable for years after completion of cisplatin-based chemotherapy (CBC). We report the largest investigation of serum platinum levels to date of 1,010 testicular cancer survivors (TCS) assessed 1-35 years after CBC and evaluate genetic contributions to these levels. EXPERIMENTAL DESIGN Eligible TCS given 300 or 400 (±15) mg/m2 cisplatin underwent extensive audiometric testing, clinical examination, completed questionnaires, and had crude serum platinum levels measured. Associations between serum platinum and various risk factors and toxicities were assessed after fitting a biexponential model adjusted for follow-up time and cumulative cisplatin dose. A genome-wide association study (GWAS) was performed using the serum platinum residuals of the dose and time-adjusted model. RESULTS Serum platinum levels exceeded the reference range for approximately 31 years, with a strong inverse relationship with creatinine clearance at follow-up (age-adjusted P = 2.13 × 10-3). We observed a significant, positive association between residual platinum values and luteinizing hormone (age-adjusted P = 6.58 × 10-3). Patients with high residual platinum levels experienced greater Raynaud phenomenon than those with medium or low levels (age-adjusted ORhigh/low = 1.46; P = 0.04), as well as a higher likelihood of developing tinnitus (age-adjusted ORhigh/low = 1.68, P = 0.07). GWAS identified one single-nucleotide polymorphism (SNP) meeting genome-wide significance, rs1377817 (P = 4.6 × 10-8, a SNP intronic to MYH14). CONCLUSIONS This study indicates that residual platinum values are correlated with several cisplatin-related toxicities. One genetic variant is associated with these levels.
Collapse
Affiliation(s)
| | - Omar El-Charif
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Mark J Ratain
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Patrick Monahan
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana
| | - Zepeng Mu
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Heather E Wheeler
- Departments of Biology and Computer Science, Loyola University Chicago, Chicago, Illinois
| | - Paul C Dinh
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana
| | - Darren R Feldman
- Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | | | - Robert J Hamilton
- Department of Surgical Oncology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - David J Vaughn
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Chunkit Fung
- J.P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | | | | | - Michiaki Kubo
- RIKEN Center for Integrative Medical Science, Yokohama, Japan
| | - Robyn Hannigan
- School for the Environment, University of Massachusetts Boston, Boston, Massachusetts
| | | | - Lawrence H Einhorn
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana
| | - Sophie D Fossa
- Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway
| | - Lois B Travis
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana.
| | - M Eileen Dolan
- Department of Medicine, University of Chicago, Chicago, Illinois.
| |
Collapse
|
120
|
Stephens M, Murphy T, Hendry D. Anaesthesia for retroperitoneal lymph node dissection in the treatment of testicular cancer. BJA Educ 2019; 19:283-289. [DOI: 10.1016/j.bjae.2019.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2019] [Indexed: 11/24/2022] Open
|
121
|
Qi L, Luo Q, Zhang Y, Jia F, Zhao Y, Wang F. Advances in Toxicological Research of the Anticancer Drug Cisplatin. Chem Res Toxicol 2019; 32:1469-1486. [PMID: 31353895 DOI: 10.1021/acs.chemrestox.9b00204] [Citation(s) in RCA: 251] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cisplatin is one of the most widely used chemotherapeutic agents for various solid tumors in the clinic due to its high efficacy and broad spectrum. The antineoplastic activity of cisplatin is mainly due to its ability to cross-link with DNA, thus blocking transcription and replication. Unfortunately, the clinical use of cisplatin is limited by its severe, dose-dependent toxic side effects. There are approximately 40 specific toxicities of cisplatin, among which nephrotoxicity is the most common one. Other common side effects include ototoxicity, neurotoxicity, gastrointestinal toxicity, hematological toxicity, cardiotoxicity, and hepatotoxicity. These side effects together reduce the life quality of patients and require lowering the dosage of the drug, even stopping administration, thus weakening the treatment effect. Few effective measures exist clinically against these side effects because the exact mechanisms of various side effects from cisplatin remain still unclear. Therefore, substantial effort has been made to explore the complicated biochemical processes involved in the toxicology of cisplatin, aiming to identify effective ways to reduce or eradicate its toxicity. This review summarizes and reviews the updated advances in the toxicological research of cisplatin. We anticipate to provide insights into the understanding of the mechanisms underlying the side effects of cisplatin and designing comprehensive therapeutic strategies involving cisplatin.
Collapse
Affiliation(s)
- Luyu Qi
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China.,University of Chinese Academy of Sciences , Beijing 100049 , P.R. China
| | - Qun Luo
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China.,University of Chinese Academy of Sciences , Beijing 100049 , P.R. China
| | - Yanyan Zhang
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China
| | - Feifei Jia
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China
| | - Yao Zhao
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China
| | - Fuyi Wang
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China.,University of Chinese Academy of Sciences , Beijing 100049 , P.R. China.,Basic Medical College , Shandong University of Chinese Traditional Medicine , Jinan 250355 , P.R. China
| |
Collapse
|
122
|
Bleyer A. Magic Still Needed for Germ Cell Tumor Research, Especially in Adolescents and Young Adults. J Oncol Pract 2019; 15:445-446. [DOI: 10.1200/jop.19.00431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Archie Bleyer
- Oregon Health and Science University, Portland, OR
- McGovern Medical School, University of Texas, Houston, TX
| |
Collapse
|
123
|
Ghandour R, Ashbrook C, Freifeld Y, Singla N, El-Asmar JM, Lotan Y, Margulis V, Eggener S, Woldu S, Bagrodia A. Nationwide Patterns of Care for Stage II Nonseminomatous Germ Cell Tumor of the Testicle. Eur Urol Oncol 2019; 3:198-206. [PMID: 31272940 DOI: 10.1016/j.euo.2019.06.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/20/2019] [Accepted: 06/11/2019] [Indexed: 11/17/2022]
Abstract
BACKGROUND Management strategies for advanced testicular cancer published from a few, high-volume clinical centers may not be generalizable. OBJECTIVE To discern treatment patterns for stage II nonseminomatous germ cell tumor (NSGCT) in a nationwide cancer registry. DESIGN, SETTING, AND PARTICIPANTS The National Cancer Database was queried for patients with a stage II NSGCT from 2004 to 2014. Patients were stratified by clinical nodal status: cN1/stage IIA, cN2/stage IIB, and cN3/stage IIIC. OUTCOMES MEASUREMENTS AND STATISTICAL ANALYSIS Logistic regression was performed to determine factors independently associated with primary retroperitoneal lymph node dissection (RPLND), primary chemotherapy, and postchemotherapy RPLND (PC-RPLND). RESULTS AND LIMITATIONS A total of 2203 patients (stages IIA, n=1060; IIB, n=869; and IIC, n=274) met the inclusion criteria. Overall, 83% of patients underwent primary chemotherapy, while 17% underwent primary RPLND. Stratified by stage, use of primary chemotherapy was 78%, 88%, and 86% for stages IIA, IIB, and IIC, respectively. Overall, 24% of patients underwent PC-RPLND. Factors independently associated with a lower likelihood of undergoing primary RPLND were a more recent diagnosis and a higher clinical nodal stage. Conversely, patients treated at high-volume facilities were more likely to receive primary RPLND. Factors associated with a higher likelihood of undergoing PC-RPLND included a higher clinical nodal stage, treatment at a high-volume center, and a greater distance of patient travel. Associations based on serum tumor markers could not be assessed. CONCLUSIONS For clinical stage II NSGCT, nationwide utilization of primary chemotherapy is increasing compared with RPLND and is the preferred therapy for more advanced nodal disease. Primary RPLND may be underutilized in stage IIA disease. Utilization of PC-RPLND is driven by nodal stage as well as accessibility of a high-volume center. PATIENT SUMMARY The use of primary retroperitoneal lymph node dissection (RPLND) in early nodal disease is declining, while upfront chemotherapy is increasingly utilized. Primary RPLND may identify patients who are actually pN0 and would not benefit from systemic chemotherapy. Primary RPLND and postchemotherapy RPLND are performed more frequently at centers of excellence.
Collapse
Affiliation(s)
- Rashed Ghandour
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Caleb Ashbrook
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yuval Freifeld
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nirmish Singla
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jose M El-Asmar
- Division of Urology, Department of Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Yair Lotan
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vitaly Margulis
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Scott Eggener
- Section of Urology, Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Solomon Woldu
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Aditya Bagrodia
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
124
|
Joly F, Ahmed-Lecheheb D, Thiery-Vuillemin A, Orillard E, Coquan E. [Side effects of chemotherapy for testicular cancers and post-cancer follow-up]. Bull Cancer 2019; 106:805-811. [PMID: 31171345 DOI: 10.1016/j.bulcan.2019.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 01/30/2023]
Abstract
Testicular cancers are the most frequent and the most curable cancers in young men. Treatments of these cancers represent a great success with cure rate over to 95 %. However, chemotherapy side effects may occur during or after several years post-treatment. This review aimed to highlight complications and physical and psychological side effects occurring mainly after chemotherapy treatment for testicular cancer, and to propose a personalized post-cancer plan specific for patients treated for testicular cancer. Treatments of these cancers can cause short-term complications (asthenia, nausea, vomiting, alopecia..). These side effects disappear within a few months after the end of the treatments. Late complications may occur several years post-treatment. Cardiovascular disease, metabolic syndrome and secondary neoplasia represent the most severe late effects among patients treated for testicular cancer. Given the increased incidence of these chemotherapy-induced side effects, it is indispensable to establish a specific follow up which must include a particular vigilance on the risk of occurrence of second cancer, a follow-up of the cardio-vascular risk factors, pulmonary and auditory follow-up, and early detection of psychosocial disorders.
Collapse
Affiliation(s)
- Florence Joly
- UNICANCER, Centre François Baclesse, Clinical Research Department and Medical Department, avenue général Harris, 14076 Caen, France; Inserm, U1086, 14076 Caen, France; Université de Caen Basse-Normandie, UMR-S1077, 14000 Caen, France; CHU de Caen, Department of Oncology, 14000 Caen, France.
| | - Djihane Ahmed-Lecheheb
- UNICANCER, Centre François Baclesse, Clinical Research Department and Medical Department, avenue général Harris, 14076 Caen, France; Inserm, U1086, 14076 Caen, France
| | | | - Emeline Orillard
- CHU Jean-Minjoz, Département Oncologie médicale, Boulevard Fleming, 25030 Besançon, France
| | - Elodie Coquan
- UNICANCER, Centre François Baclesse, Clinical Research Department and Medical Department, avenue général Harris, 14076 Caen, France
| |
Collapse
|
125
|
Indications, evolving technique, and early outcomes with robotic retroperitoneal lymph node dissection. Curr Opin Urol 2019; 28:461-468. [PMID: 29979235 DOI: 10.1097/mou.0000000000000530] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW Examine and discuss indications, technique, and outcomes for robotic retroperitoneal lymph node dissection (RPLND) for testicular cancer. RECENT FINDINGS Open RPLND has been the longstanding standard of care for both primary and post chemotherapy RPLND. Recently, robotic RPLND has been an attractive option with the intent of reducing the morbidity associated with open surgery while providing identical oncologic efficacy. Naysayers of robotic RPLND suggest it is often inappropriately used as a staging procedure and consequently can compromise oncologic efficacy. SUMMARY Robotic RPLND is being evaluated as a therapeutic equivalent to open RPLND. On the basis of limited published data with modest follow-up from experienced centers, robotic RPLND appears to provide effective staging and therapeutic data mirroring that of open surgery.
Collapse
|
126
|
Hamilton RJ, Jewett MAS, Warde P, Hansen A. Optimal Management of High-risk Stage I Nonseminomatous Germ Cell Tumor: Surveillance is the Preferred Option. Eur Urol Focus 2019; 5:702-703. [PMID: 31129064 DOI: 10.1016/j.euf.2019.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 05/03/2019] [Indexed: 11/25/2022]
Abstract
At Princess Margaret, we recommend active surveillance for all patients with clinical stage I nonseminomatous germ cell tumor. Here we refute common arguments against surveillance and urge clinicians to engage in a shared decision-making process that goes beyond merely citing relapse rates for the different options.
Collapse
Affiliation(s)
- Robert J Hamilton
- Department of Surgery (Urology), Princess Margaret Cancer Centre, University Health Network and The University of Toronto, Toronto, Canada.
| | - Michael A S Jewett
- Department of Surgery (Urology), Princess Margaret Cancer Centre, University Health Network and The University of Toronto, Toronto, Canada
| | - Padraig Warde
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Aaron Hansen
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| |
Collapse
|
127
|
Makharza SA, Cirillo G, Vittorio O, Valli E, Voli F, Farfalla A, Curcio M, Iemma F, Nicoletta FP, El-Gendy AA, Goya GF, Hampel S. Magnetic Graphene Oxide Nanocarrier for Targeted Delivery of Cisplatin: A Perspective for Glioblastoma Treatment. Pharmaceuticals (Basel) 2019; 12:E76. [PMID: 31109098 PMCID: PMC6631527 DOI: 10.3390/ph12020076] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/10/2019] [Accepted: 05/16/2019] [Indexed: 12/19/2022] Open
Abstract
Selective vectorization of Cisplatin (CisPt) to Glioblastoma U87 cells was exploited by the fabrication of a hybrid nanocarrier composed of magnetic γ-Fe2O3 nanoparticles and nanographene oxide (NGO). The magnetic component, obtained by annealing magnetite Fe3O4 and characterized by XRD measurements, was combined with NGO sheets prepared via a modified Hummer's method. The morphological and thermogravimetric analysis proved the effective binding of γ-Fe2O3 nanoparticles onto NGO layers. The magnetization measured under magnetic fields up to 7 Tesla at room temperature revealed superparamagnetic-like behavior with a maximum value of MS = 15 emu/g and coercivity HC ≈ 0 Oe within experimental error. The nanohybrid was found to possess high affinity towards CisPt, and a rather slow fractional release profile of 80% after 250 h. Negligible toxicity was observed for empty nanoparticles, while the retainment of CisPt anticancer activity upon loading into the carrier was observed, together with the possibility to spatially control the drug delivery at a target site.
Collapse
Affiliation(s)
- Sami A Makharza
- Leibniz Institute of Solid State and Material Research Dresden, 01069 Dresden, Germany.
- College of Pharmacy and Medical Sciences, Hebron University, Hebron 00970, Palestine.
| | - Giuseppe Cirillo
- Leibniz Institute of Solid State and Material Research Dresden, 01069 Dresden, Germany.
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), 87036 Rende, Italy.
| | - Orazio Vittorio
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2031, Australia.
- ARC Centre of Excellence for Convergent BioNano Science and Technology, Australian Centre for NanoMedicine, UNSW Sydney, Sydney 2052, Australia.
- School of Women's and Children's Health, Faculty of Medicine, UNSW Sydney, Sydney 2052, Australia.
| | - Emanuele Valli
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2031, Australia.
- School of Women's and Children's Health, Faculty of Medicine, UNSW Sydney, Sydney 2052, Australia.
| | - Florida Voli
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2031, Australia.
| | - Annafranca Farfalla
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), 87036 Rende, Italy.
| | - Manuela Curcio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), 87036 Rende, Italy.
| | - Francesca Iemma
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), 87036 Rende, Italy.
| | - Fiore Pasquale Nicoletta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), 87036 Rende, Italy.
| | - Ahmed A El-Gendy
- Department of Physics, University of Texas at El Paso, El Paso, TX 79968, USA.
| | - Gerardo F Goya
- Institute of Nanoscience of Aragon (INA) & Department of Condensed Matter Physics, University of Zaragoza, 50018 Zaragoza, Spain.
| | - Silke Hampel
- Leibniz Institute of Solid State and Material Research Dresden, 01069 Dresden, Germany.
| |
Collapse
|
128
|
Zazuli Z, Otten LS, Drögemöller BI, Medeiros M, Monzon JG, Wright GEB, Kollmannsberger CK, Bedard PL, Chen Z, Gelmon KA, McGoldrick N, Kitchlu A, Vijverberg SJH, Masereeuw R, Ross CJD, Liu G, Carleton BC, Maitland-van der Zee AH. Outcome Definition Influences the Relationship Between Genetic Polymorphisms of ERCC1, ERCC2, SLC22A2 and Cisplatin Nephrotoxicity in Adult Testicular Cancer Patients. Genes (Basel) 2019; 10:E364. [PMID: 31083486 PMCID: PMC6562793 DOI: 10.3390/genes10050364] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 05/07/2019] [Indexed: 12/16/2022] Open
Abstract
Although previous research identified candidate genetic polymorphisms associated with cisplatin nephrotoxicity, varying outcome definitions potentially contributed to the variability in the effect size and direction of this relationship. We selected genetic variants that have been significantly associated with cisplatin-induced nephrotoxicity in more than one published study (SLC22A2 rs316019; ERCC1 rs11615 and rs3212986; ERCC2 rs1799793 and rs13181) and performed a replication analysis to confirm associations between these genetic polymorphisms and cisplatin nephrotoxicity using various outcome definitions. We included 282 germ cell testicular cancer patients treated with cisplatin from 2009-2014, aged >17 years recruited by the Canadian Pharmacogenomics Network for Drug Safety. Nephrotoxicity was defined using four grading tools: (1) Common Terminology Criteria for Adverse Events (CTCAE) v4.03 for acute kidney injury (AKI) or CTCAE-AKI; (2) adjusted cisplatin-induced AKI; (3) elevation of serum creatinine; and (4) reduction in the estimated glomerular filtration rate (eGFR). Significant associations were only found when using the CTCAE v4.03 definition: genotype CA of the ERCC1 rs3212986 was associated with decreased risk of cisplatin nephrotoxicity (ORadj = 0.24; 95% CI:0.08-0.70; p= 0.009) compared to genotype CC. In contrast, addition of allele A at SLC22A2 rs316019 was associated with increased risk (ORadj = 4.41; 95% CI:1.96-9.88; p < 0.001) while genotype AC was associated with a higher risk of cisplatin nephrotoxicity (ORadj = 5.06; 95% CI:1.69-15.16; p= 0.004) compared to genotype CC. Our study showed that different case definitions led to variability in the genetic risk ascertainment of cisplatin nephrotoxicity. Therefore, consensus on a set of clinically relevant outcome definitions that all such studies should follow is needed.
Collapse
Affiliation(s)
- Zulfan Zazuli
- Department of Respiratory Medicine, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
- Department of Pharmacology-Clinical Pharmacy, School of Pharmacy, Bandung Institute of Technology, Bandung 40132, Indonesia.
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3512 JE Utrecht, The Netherlands.
| | - Leila S Otten
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3512 JE Utrecht, The Netherlands.
| | - Britt I Drögemöller
- British Columbia Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada.
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| | - Mara Medeiros
- Nephrology Research Unit, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico.
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico.
| | - Jose G Monzon
- Department of Medical Oncology, Tom Baker Cancer Centre, Calgary, AB T2N 4N2, Canada.
| | - Galen E B Wright
- British Columbia Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| | | | - Philippe L Bedard
- Princess Margaret Cancer Centre and University of Toronto, Toronto, ON M5S, Canada.
| | - Zhuo Chen
- Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre-University Health Network and University of Toronto, Toronto, ON M5S, Canada.
| | - Karen A Gelmon
- BC Cancer Agency and University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| | - Nicole McGoldrick
- Pharmaceutical Outcomes Programme, BC Children's Hospital, Vancouver, BC V6H 3N1, Canada.
| | - Abhijat Kitchlu
- Division of Nephrology, Department of Medicine, University Health Network and University of Toronto, Toronto, ON M5S, Canada.
| | - Susanne J H Vijverberg
- Department of Respiratory Medicine, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3512 JE Utrecht, The Netherlands.
| | - Colin J D Ross
- British Columbia Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada.
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| | - Geoffrey Liu
- Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre-University Health Network and University of Toronto, Toronto, ON M5S, Canada.
| | - Bruce C Carleton
- Pharmaceutical Outcomes Programme, BC Children's Hospital, Vancouver, BC V6H 3N1, Canada.
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| | - Anke H Maitland-van der Zee
- Department of Respiratory Medicine, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
129
|
Abstract
PURPOSE OF REVIEW In the present review, we summarize the recent developments in the management of germ cell tumors (GCTs). RECENT FINDINGS Treatment-related acute and late-onset toxicity remains a key challenge in the management of GCTs, with recent evidence showing that the adverse health outcomes of etoposide and cisplatin for four cycles in comparison to bleomycin, etoposide, and cisplatin for three cycles appear to be similar. Recent data showed that multidisciplinary clinic approach and management in experienced academic centers were associated with improved overall survival in GCT patients. There are currently multiple conventional-dose chemotherapy options for salvage therapy in patients with refractory or recurrent disease. In addition, more efficacious high-dose chemotherapy regimens continue to be developed. The role of salvage conventional-dose chemotherapy versus high-dose chemotherapy is currently being investigated prospectively. Recent reports suggested that brentuximab vedotin could be a potential salvage option for cluster of differentiation 30 positive refractory GCTs. On the other hand the results of the first phase II clinical trial investigating pembrolizumab in refractory GCTs were disappointing showing no clinical activity.Finally, deep exploration of the immune profile of GCTs using immunohistochemistry and gene expression profiling has identified that advanced GCT stage was associated with decreased T-cell and Natural killer-cell signatures, whereas T regulatory, neutrophil, mast cell, and macrophage signatures increased with advanced stage. Even though these results indicated that activated T-cell infiltration correlated with seminoma histology and good prognosis, and could be used in the future as a biomarker, this approach needs to be validated in a large cohort. SUMMARY Remaining challenges to be addressed include minimizing therapeutic toxicity, and improving outcomes in patients with refractory/recurrent GCTs.
Collapse
|
130
|
Shankara-Narayana N, Di Pierro I, Fennell C, Ly LP, Bacha F, Vrga L, Savkovic S, Turner L, Jayadev V, Conway AJ, Handelsman DJ. Sperm cryopreservation prior to gonadotoxic treatment: experience of a single academic centre over 4 decades. Hum Reprod 2019; 34:795-803. [DOI: 10.1093/humrep/dez026] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/23/2019] [Indexed: 01/19/2023] Open
Affiliation(s)
- Nandini Shankara-Narayana
- Andrology Department, Concord Hospital, Hospital Road, Concord Hospital, New South Wales, Australia
- ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - Irene Di Pierro
- Andrology Department, Concord Hospital, Hospital Road, Concord Hospital, New South Wales, Australia
| | - Carolyn Fennell
- Andrology Department, Concord Hospital, Hospital Road, Concord Hospital, New South Wales, Australia
| | - Lam P Ly
- Andrology Department, Concord Hospital, Hospital Road, Concord Hospital, New South Wales, Australia
- ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - Fay Bacha
- Andrology Department, Concord Hospital, Hospital Road, Concord Hospital, New South Wales, Australia
| | - Ljubica Vrga
- Andrology Department, Concord Hospital, Hospital Road, Concord Hospital, New South Wales, Australia
| | - Sasha Savkovic
- Andrology Department, Concord Hospital, Hospital Road, Concord Hospital, New South Wales, Australia
| | - Leo Turner
- Andrology Department, Concord Hospital, Hospital Road, Concord Hospital, New South Wales, Australia
| | - Veena Jayadev
- Andrology Department, Concord Hospital, Hospital Road, Concord Hospital, New South Wales, Australia
| | - Ann J Conway
- Andrology Department, Concord Hospital, Hospital Road, Concord Hospital, New South Wales, Australia
- ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - David J Handelsman
- Andrology Department, Concord Hospital, Hospital Road, Concord Hospital, New South Wales, Australia
- ANZAC Research Institute, University of Sydney, Sydney, Australia
| |
Collapse
|
131
|
Second cancers and causes of death in patients with testicular cancer in Sweden. PLoS One 2019; 14:e0214410. [PMID: 30921367 PMCID: PMC6438485 DOI: 10.1371/journal.pone.0214410] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/12/2019] [Indexed: 12/15/2022] Open
Abstract
While treatment for testicular cancer (TC) has become standardized after the 1980s with an associated significant improvement in patient survival, this has been accompanied by an increased risk of second primary cancers (SPCs). Patients were identified from the Swedish Cancer Registry spanning the years from 1980 to 2015, including 8788 individuals with primary TC and their SPCs. Relative risks (RRs) for SPC were calculated using the generalized Poisson regression model. SPCs were diagnosed in 9.4% of patients with TC and half of them were late onset cancers not common in the population in their 40s. Overall RR of SPCs (excluding second TC) was 1.30 (95%CI: 1.20–1.40), including high risks for seven solid cancers, non-Hodgkin lymphoma and leukemia. Second TC was the most common SPC and the RR of 17.19 (95%CI: 14.89–19.85) was the highest recorded. Cancers known to be fatal as first primary cancers were also fatal as SPC in TC patients. Survival at 30 years of follow-up was approximately 80% for TC patients without SPC but it decreased to 40% for patients with SPC. The unexpected finding that half of the identified SPCs were typical late onset cancers in the middle-aged population raises concerns that therapy may facilitate premature aging. The risks of SPC are clinically important for the long-term management of TC patients and the high-mortality calls for a future management strategy.
Collapse
|
132
|
Alsdorf W, Seidel C, Bokemeyer C, Oing C. Current pharmacotherapy for testicular germ cell cancer. Expert Opin Pharmacother 2019; 20:837-850. [PMID: 30849243 DOI: 10.1080/14656566.2019.1583745] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION With the implementation of platinum-based chemotherapy, germ cell tumors (GCTs) became a model for a curable solid tumor, with survival rates of 95% in all patients with >80% survival in metastatic stages. AREAS COVERED Herein, the authors review the current standards of adjuvant chemotherapy for stage I GCTs as well as first-line and salvage treatments for metastatic disease. Novel approaches for refractory disease are also reviewed. EXPERT OPINION Active surveillance should be considered for all stage I patients and is the preferred approach for stage I seminoma. In stage I non-seminomas with vascular invasion, one cycle of bleomycin, etoposide, and cisplatin (BEP) substantially reduces the relapse risk. For most advanced GCTs, BEP remains the first-line standard of care. For poor prognosis disease treatment, stratification according to tumor marker decline is recommended. The role of primary high-dose chemotherapy (HDCT) for selected very high-risk patients remains to be prospectively evaluated. Salvage HDCT at relapse seems superior to conventional chemotherapy, retrospectively. The treatment of multiply relapsed disease remains challenging. The gemcitabine/oxaliplatin/paclitaxel (GOP) protocol is considered the standard for refractory disease. However, overall, outcomes are poor and new treatment approaches are urgently needed with targeted therapies so far failing to yield relevant clinical activity.
Collapse
Affiliation(s)
- Winfried Alsdorf
- a Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology , University Medical Center Eppendorf , Hamburg , Germany
| | - Christoph Seidel
- a Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology , University Medical Center Eppendorf , Hamburg , Germany
| | - Carsten Bokemeyer
- a Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology , University Medical Center Eppendorf , Hamburg , Germany
| | - Christoph Oing
- a Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology , University Medical Center Eppendorf , Hamburg , Germany.,b Laboratory of Radiobiology and Experimental Radiation Oncology , University Medical Center Eppendorf , Hamburg , Germany
| |
Collapse
|
133
|
|
134
|
Fankhauser CD, Gerke TA, Roth L, Sander S, Grossmann NC, Kranzbühler B, Eberli D, Sulser T, Beyer J, Hermanns T. Pre-orchiectomy tumor marker levels should not be used for International Germ Cell Consensus Classification (IGCCCG) risk group assignment. J Cancer Res Clin Oncol 2019; 145:781-785. [PMID: 30637464 DOI: 10.1007/s00432-019-02844-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 01/08/2019] [Indexed: 01/17/2023]
Abstract
PURPOSE To investigate whether the use of pre-orchiectomy instead of pre-chemotherapy tumor marker (TM) levels has an impact on the International Germ Cell Consensus Classification (IGCCCG) risk group assignment in patients with metastatic germ cell tumors (GCT). METHODS Demographic and clinical information of all patients treated for primary metastatic testicular non-seminomatous GCT in our tertiary care academic center were extracted from medical charts. IGCCCG risk group assignment was correctly performed with pre-chemotherapy marker levels and additionally with pre-orchiectomy marker levels. Agreement between pre-chemotherapy and pre-orchiectomy risk group assignments was assessed using Cohen's kappa. RESULTS Our cohort consisted of 83 patients. The use of pre-orchiectomy TMs resulted in an IGCCCG risk group upstaging in 12 patients (16%, 8 patients from good to intermediate risk and 4 patients from intermediate to poor risk) and a downstaging in 1 patient (1.2%, from intermediate- to good-risk). The agreement between pre-orchiectomy and pre-chemotherapy IGCCCG risk groups resulted in a Cohen's kappa of 0.888 (p < 0.001). CONCLUSIONS Using pre-orchiectomy TMs can result in incorrect IGCCCG risk group assignment, which in turn can impact on the clinical management and follow-up of patients with metastatic GCT. Thus, adherence to the IGCCCG standard using pre-chemotherapy TMs levels is recommended.
Collapse
Affiliation(s)
| | - Travis A Gerke
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, USA
| | - Lisa Roth
- Department of Urology, University Hospital, University of Zurich, Zurich, Switzerland
| | - Sophia Sander
- Department of Urology, University Hospital, University of Zurich, Zurich, Switzerland
| | | | - Benedikt Kranzbühler
- Department of Urology, University Hospital, University of Zurich, Zurich, Switzerland
| | - Daniel Eberli
- Department of Urology, University Hospital, University of Zurich, Zurich, Switzerland
| | - Tullio Sulser
- Department of Urology, University Hospital, University of Zurich, Zurich, Switzerland
| | - Joerg Beyer
- Department of Medical Oncology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland.
| | - Thomas Hermanns
- Department of Urology, University Hospital, University of Zurich, Zurich, Switzerland
| |
Collapse
|
135
|
Panza S, Gelsomino L, Malivindi R, Rago V, Barone I, Giordano C, Giordano F, Leggio A, Comandè A, Liguori A, Aquila S, Bonofiglio D, Andò S, Catalano S. Leptin Receptor as a Potential Target to Inhibit Human Testicular Seminoma Growth. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:687-698. [PMID: 30610844 DOI: 10.1016/j.ajpath.2018.11.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 11/07/2018] [Accepted: 11/16/2018] [Indexed: 12/11/2022]
Abstract
Although in past decades the adipokine leptin and its own receptor have been considered as significant cancer biomarkers, their potential involvement in human testicular seminoma growth and progression remains unexplored. Here, we showed that the expression of leptin and its receptor was significantly higher in human testicular seminoma compared with normal adult testis. Human seminoma cell line TCam-2 also expressed leptin along with the long and short isoforms of leptin receptor, and in response to leptin treatment showed enhanced activation of its downstream effectors. In line with these results, leptin stimulation significantly increased the proliferation and migration of TCam-2 cells. Treatment of TCam-2 cells with the peptide Leu-Asp-Phe-Ile (LDFI), a full leptin-receptor antagonist, completely reversed the leptin-mediated effects on cell growth and motility as well as reduced the expression of several leptin-induced target genes. More importantly, the in vivo xenograft experiments showed that LDFI treatment markedly decreased seminoma tumor growth. Interestingly, LDFI-treated tumors showed reduced levels of the proliferation marker Ki-67 as well as decreased expression of leptin-regulated genes. Taken together, these data identify, for the first time, leptin as a key factor able to affect testicular seminoma behavior, highlighting leptin receptor as a potential target for novel potential treatments in this type of cancer.
Collapse
Affiliation(s)
- Salvatore Panza
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Rocco Malivindi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Vittoria Rago
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | | | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Antonella Leggio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Alessandra Comandè
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Angelo Liguori
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Saveria Aquila
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| |
Collapse
|
136
|
Essex AL, Pin F, Huot JR, Bonewald LF, Plotkin LI, Bonetto A. Bisphosphonate Treatment Ameliorates Chemotherapy-Induced Bone and Muscle Abnormalities in Young Mice. Front Endocrinol (Lausanne) 2019; 10:809. [PMID: 31803146 PMCID: PMC6877551 DOI: 10.3389/fendo.2019.00809] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 11/04/2019] [Indexed: 12/14/2022] Open
Abstract
Chemotherapy is frequently accompanied by several side effects, including nausea, diarrhea, anorexia and fatigue. Evidence from ours and other groups suggests that chemotherapy can also play a major role in causing not only cachexia, but also bone loss. This complicates prognosis and survival among cancer patients, affects quality of life, and can increase morbidity and mortality rates. Recent findings suggest that soluble factors released from resorbing bone directly contribute to loss of muscle mass and function secondary to metastatic cancer. However, it remains unknown whether similar mechanisms also take place following treatments with anticancer drugs. In this study, we found that young male CD2F1 mice (8-week old) treated with the chemotherapeutic agent cisplatin (2.5 mg/kg) presented marked loss of muscle and bone mass. Myotubes exposed to bone conditioned medium from cisplatin-treated mice showed severe atrophy (-33%) suggesting a bone to muscle crosstalk. To test this hypothesis, mice were administered cisplatin in combination with an antiresorptive drug to determine if preservation of bone mass has an effect on muscle mass and strength following chemotherapy treatment. Mice received cisplatin alone or combined with zoledronic acid (ZA; 5 μg/kg), a bisphosphonate routinely used for the treatment of osteoporosis. We found that cisplatin resulted in progressive loss of body weight (-25%), in line with reduced fat (-58%) and lean (-17%) mass. As expected, microCT bone histomorphometry analysis revealed significant reduction in bone mass following administration of chemotherapy, in line with reduced trabecular bone volume (BV/TV) and number (Tb.N), as well as increased trabecular separation (Tb.Sp) in the distal femur. Conversely, trabecular bone was protected when cisplatin was administered in combination with ZA. Interestingly, while the animals exposed to chemotherapy presented significant muscle wasting (~-20% vs. vehicle-treated mice), the administration of ZA in combination with cisplatin resulted in preservation of muscle mass (+12%) and strength (+42%). Altogether, these observations support our hypothesis of bone factors targeting muscle and suggest that pharmacological preservation of bone mass can benefit muscle mass and function following chemotherapy.
Collapse
Affiliation(s)
- Alyson L. Essex
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Fabrizio Pin
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Joshua R. Huot
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lynda F. Bonewald
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
- Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, United States
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- IUPUI Center for Cachexia Research, Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lilian I. Plotkin
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Andrea Bonetto
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
- Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, United States
- IUPUI Center for Cachexia Research, Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Otolaryngology – Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- *Correspondence: Andrea Bonetto
| |
Collapse
|
137
|
Tedesco KT, Sarthy J, Pinto N, Boos MD. Acute enlargement of a vascular plaque and gait changes in a young girl. BMJ 2018; 363:k4679. [PMID: 30487229 DOI: 10.1136/bmj.k4679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
| | - Jay Sarthy
- Division of Hematology-Oncology, Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital; Seattle, WA, USA
| | - Navin Pinto
- Division of Hematology-Oncology, Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital; Seattle, WA, USA
| | - Markus D Boos
- Division of Dermatology, Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital; Seattle, WA, USA
| |
Collapse
|
138
|
Dieckmann KP, Anheuser P, Kulejewski M, Gehrckens R, Feyerabend B. Is there still a place for retroperitoneal lymph node dissection in clinical stage 1 nonseminomatous testicular germ-cell tumours? A retrospective clinical study. BMC Urol 2018; 18:95. [PMID: 30367648 PMCID: PMC6204050 DOI: 10.1186/s12894-018-0412-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 10/17/2018] [Indexed: 01/22/2023] Open
Abstract
Background Primary retroperitoneal lymph node dissection (RPLND) ultimately lost its role as the standard management of clinical stage (CS) 1 nonseminomatous (NS) testicular germ cell tumours (GCTs) in Europe when the European Germ Cell Cancer Consensus Group released their recommendations in 2008. Current guide-lines recommend surgery only for selected patients but reasons for selection remain rather ill-defined. We evaluated the practice patterns of the management of CS1 patients and looked specifically to the role of RPLND among other standard treatment options. Methods We retrospectively evaluated the treatment modalities of 75 consecutive patients treated for CS1 NS at one centre during 2008–2017. The patients undergoing RPLND were selected for a closer review. Particular reasons for surgery, clinical features of patients, and therapeutic outcome were analyzed using descriptive statistical methods. Results Twelve patients (16%) underwent nerve-sparing RPLND, nine surveillance, 54 had various regimens of adjuvant chemotherapy. Particular reasons for surgery involved illnesses precluding chemotherapy (n = 2), patients´ choice (n = 4), and teratomatous histology of the primary associated with equivocal radiologic findings (n = 6). Five patients had lymph node metastases, two received additional chemotherapy. Antegrade ejaculation was preserved in all cases. One patient had a grade 2 complication that was managed conservatively. All RPLND-patients remained disease-free. Conclusions Primary RPLND is a useful option in distinct CS1 patients, notably those with concurrent health problems precluding chemotherapy, and those with high proportions of teratoma in the primary associated with equivocal radiological findings. Informed patient’s preference represents another acceptable reason for the procedure. RPLND properly suits the needs of well-selected patients with CS1 nonseminoma and deserves consideration upon clinical decision-making.
Collapse
Affiliation(s)
- K-P Dieckmann
- Albertinen-Krankenhaus Hamburg, Klinik für Urologie, Hamburg, Germany. .,Asklepios Klinik Altona, Urologische Abteilung, Hodentumorzentrum Hamburg, Hamburg, Germany. .,Asklepios Klinik Altona, Hodentumorzentrum Hamburg, Paul Ehrlich Strasse 1, 22763, Hamburg, Germany.
| | - P Anheuser
- Albertinen-Krankenhaus Hamburg, Klinik für Urologie, Hamburg, Germany
| | - M Kulejewski
- Albertinen-Krankenhaus Hamburg, Klinik für Urologie, Hamburg, Germany
| | - R Gehrckens
- Albertinen-Krankenhaus Hamburg, Klinik für Diagnostische Radiologie, Hamburg, Germany
| | | |
Collapse
|
139
|
Terenziani M, De Pasquale MD, Bisogno G, Biasoni D, Boldrini R, Collini P, Conte M, Dall'Igna P, Inserra A, Melchionda F, Siracusa F, Spreafico F, Barretta F, D'Angelo P. Malignant testicular germ cell tumors in children and adolescents: The AIEOP (Associazione Italiana Ematologia Oncologia Pediatrica) protocol. Urol Oncol 2018; 36:502.e7-502.e13. [PMID: 30249520 DOI: 10.1016/j.urolonc.2018.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/25/2018] [Accepted: 07/02/2018] [Indexed: 11/19/2022]
Abstract
OBJECTIVES We report the results of an Associazione Italiana Ematologia Oncologia Pediatrica (AIEOP) study on the treatment of testicular germ cell tumors (TGCT) with a pediatric PEB (pPEB) regimen (cisplatin 25 mg/m2 daily on days 1-4; etoposide 100 mg/m2 daily on days 1-4; bleomycin 15 mg/m2 on day 2, once per cycle). METHODS AND MATERIALS Male patients under 18 years old with malignant TGCT were enrolled for a second national prospective protocol. All patients underwent orchiectomy at diagnosis. Those with Stage I received no chemotherapy; those with Stage II-III disease received three cycles of pPEB; and those with Stage IV received four cycles. After chemotherapy, resection of radiologically-evident residual disease was recommended. The main study end-points were overall survival and relapse-free survival. RESULTS Ninety-nine boys from 0.5 to 17.8 years old (median 15.4 years) were evaluable, and staged as follows: 58 Stage I (59%), 7 Stage II (7%), 14 Stage III (14%), and 20 Stage IV (20%). With a median follow-up of 59 months (range 4-165 months), 5-year relapse-free survival (95% CI) was 73% (65%-83%) for the whole sample, 65% (53%-79%) for Stage I patients, and 86% (75%-98%) for Stage II-IV patients. Five-year overall survival (95% CI) was 99% (97%-100%). CONCLUSIONS We confirmed a good prognosis for malignant TGCT in children and adolescents. Reducing the number of chemotherapy cycles for Stage II-III disease does not seem to negatively affect survival outcomes.
Collapse
Affiliation(s)
- Monica Terenziani
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| | - Maria D De Pasquale
- Hematology/Oncology Department, Ospedale Pediatrico Bambino Gesù-IRCCS, Roma, Italy
| | - Gianni Bisogno
- Pediatric Unit, University-Hospital of Padua, Padova, Italy
| | - Davide Biasoni
- Pediatric Surgery Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Renata Boldrini
- Pathology Unit, Ospedale Pediatrico Bambino Gesù-IRCCS, Roma, Italy
| | - Paola Collini
- Pathology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Massimo Conte
- Oncology Unit, Ospedale Pediatrico G. Gaslini, Genova, Italy
| | - Patrizia Dall'Igna
- Pediatric Surgery Department, University-Hospital of Padua, Padova, Italy
| | - Alessandro Inserra
- Pediatric Surgery Department, Ospedale Pediatrico Bambino Gesù-IRCCS, Roma, Italy
| | | | | | - Filippo Spreafico
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Francesco Barretta
- Unit of Clinical Epidemiology and Trial Organization, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Paolo D'Angelo
- Hematology/Oncology Unit, A.R.N.A.S Civico Di Cristina e Benfratelli, Palermo, Italy
| |
Collapse
|
140
|
Sonpavde GP, Mariani L, Lo Vullo S, Raggi D, Giannatempo P, Bamias A, Crabb SJ, Bellmunt J, Yu EY, Niegisch G, Vaishampayan UN, Theodore C, Berthold DR, Srinivas S, Sridhar SS, Plimack ER, Rosenberg JE, Powles T, Galsky MD, Necchi A. Impact of the Number of Cycles of Platinum Based First Line Chemotherapy for Advanced Urothelial Carcinoma. J Urol 2018; 200:1207-1214. [PMID: 30012366 DOI: 10.1016/j.juro.2018.07.035] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2018] [Indexed: 12/20/2022]
Abstract
PURPOSE We evaluated the impact of the number of cycles of platinum based, first line chemotherapy (fewer than 6 cycles vs the conventional 6 cycles or more) on the survival of patients with metastatic urothelial carcinoma. MATERIALS AND METHODS We used the RISC (Retrospective International Study of Invasive/Advanced Cancer of the Urothelium) database. The association of the number of cycles of chemotherapy with overall survival was investigated by Cox multiple regression analysis after controlling for recognized prognostic factors. We excluded patients who received fewer than 3 or more than 9 platinum chemotherapy cycles to reduce confounding factors. The primary analysis was a comparison of overall survival for 3 to 5 vs 6 to 9 cycles using 6-month landmark analysis when 281 death events were observed. RESULTS Of the 1,020 patients in the RISC 472 received cisplatin or carboplatin, of whom 338 and 134, respectively, were evaluable. A total of 157 patients received 3 to 5 cycles (median 4) and 315 received 6 to 9 cycles (median 6). There was no significant difference in overall survival between 3 to 5 and 6 to 9 cycles (HR 1.02, 95% CI 0.78-1.33, p = 0.91). No significant interactions were observed for the type of platinum (p = 0.09) and completed planned chemotherapy (p = 0.56). The limitations of a hypothesis generating, retrospective analysis applied. CONCLUSIONS Four cycles of platinum based, first line chemotherapy appeared adequate and did not significantly compromise the survival of patients with advanced urothelial carcinoma. The omission of excessive cycles may avoid unnecessary cumulative toxicity and facilitate a better transition to second line therapy and investigational switch maintenance therapy strategies. These results require prospective validation but they may impact practice in select patients.
Collapse
Affiliation(s)
| | - Luigi Mariani
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Daniele Raggi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | | | | | - Evan Y Yu
- University of Washington, Seattle, Washington
| | - Guenter Niegisch
- Department of Urology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | | | | | | | | | | | | | | | - Thomas Powles
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Matthew D Galsky
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, New York
| | - Andrea Necchi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
141
|
Intermediate and long-term complications associated with adjuvant chemotherapy for stage I germ cell tumor patients. Curr Opin Urol 2018; 28:485-490. [DOI: 10.1097/mou.0000000000000527] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
142
|
Maroto P, Anguera G, Martin C. Long-term toxicity of the treatment for germ cell-cancer. A review. Crit Rev Oncol Hematol 2018; 121:62-67. [DOI: 10.1016/j.critrevonc.2017.11.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 11/07/2017] [Accepted: 11/20/2017] [Indexed: 12/18/2022] Open
|