1
|
Aujla P, Aleem AS, Khanna R, Mittal N, Lyons J. Metastatic Pure Choriocarcinoma Masquerading in the Gastrointestinal Tract. Cureus 2024; 16:e64566. [PMID: 39144869 PMCID: PMC11323720 DOI: 10.7759/cureus.64566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
This study presents a rare case of pure choriocarcinoma (PCC) with metastasis to the gastrointestinal tract in a 52-year-old male with a history of mixed germ cell tumor in remission. Despite negative oncology surveillance imaging, serum marker monitoring, and a recent colonoscopy, the patient presented with new-onset melena and dysphagia, leading to further diagnostic evaluation. Endoscopic examination revealed an ulcerated duodenal mass, and a computer tomography (CT)-guided liver biopsy confirmed metastatic PCC. This case highlights the aggressive nature of PCC and the importance of considering gastrointestinal metastasis in patients with atypical symptoms, even when in apparent remission.
Collapse
Affiliation(s)
- Parvir Aujla
- Internal Medicine, University of Texas Health Science Center at Houston, Houston, USA
| | - Abdullah S Aleem
- Internal Medicine, University of Texas Health Science Center at Houston, Houston, USA
| | - Rohit Khanna
- Gastroenterology and Hepatology, Scripps Clinic, La Jolla, USA
| | - Nitish Mittal
- Internal Medicine, University of Texas Health Science Center at Houston, Houston, USA
| | - John Lyons
- Gastroenterology and Hepatology, Scripps Clinic, La Jolla, USA
| |
Collapse
|
2
|
Kirisawa T, Okuno T, Hagimoto H, Matsuda A, Maejima A, Shinoda Y, Nakamura E, Komiyama M, Fujimoto H, Matsui Y. Clinical characteristics and survival outcomes of elderly patients with de novo metastatic germ cell tumors. Int J Urol 2024; 31:804-812. [PMID: 38693651 DOI: 10.1111/iju.15470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/03/2024] [Indexed: 05/03/2024]
Abstract
OBJECTIVES To determine the outcomes for elderly patients with de novo metastatic germ cell tumors and the influence of patient age on adherence to standard chemotherapy. METHODS A total of 150 patients who were initially diagnosed with metastatic germ cell tumors and treated at our institution between 2007 and 2021 were included. Patients were classified according to three age groups: aged <40, 40-49, and ≥50 years. Clinicopathological features, adherence to standard first-line chemotherapy, overall survival, and disease-free survival were compared between these groups. We also analyzed the outcomes of patients who received low-intensity induction chemotherapy due to adverse events and/or comorbidities. RESULTS There was no significant difference in any of the survival outcomes and in the rate of adherence to standard first-line chemotherapy between the three age groups, although elderly patients with intermediate/poor prognosis group tended to receive less-intense chemotherapies. The rate of febrile neutropenia as a chemotherapy-related adverse event was significantly higher in patients aged ≥50 years. No statistical significance in survival outcomes was detected between the group of patients who received relatively low-intensity induction chemotherapy and those who received adequately intensive planned chemotherapy. CONCLUSIONS The adherence rate of standard fist-line chemotherapy of elderly patients is almost comparable to that of younger patients, although some adverse events should be carefully managed. Even elderly patients with metastatic germ cell tumors can aim for equivalently good survival outcome like younger populations, with effort to adhere to standard chemotherapy.
Collapse
Affiliation(s)
- Takahiro Kirisawa
- Department of Urology and Retroperitoneal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Tomoya Okuno
- Department of Urology and Retroperitoneal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroki Hagimoto
- Department of Urology and Retroperitoneal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Ayumu Matsuda
- Department of Urology and Retroperitoneal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Aiko Maejima
- Department of Urology and Retroperitoneal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Yasuo Shinoda
- Department of Urology and Retroperitoneal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Eijiro Nakamura
- Department of Urology and Retroperitoneal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Motokiyo Komiyama
- Department of Urology and Retroperitoneal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroyuki Fujimoto
- Department of Urology and Retroperitoneal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Yoshiyuki Matsui
- Department of Urology and Retroperitoneal Surgery, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
3
|
Kobayashi K, Kishida T. Predictors of the development of febrile neutropenia in chemotherapy for advanced germ cell tumors. Int J Urol 2024; 31:134-138. [PMID: 37915108 DOI: 10.1111/iju.15323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 09/26/2023] [Indexed: 11/03/2023]
Abstract
OBJECTIVES To identify the predictive factors for the development of febrile neutropenia (FN) in the course of chemotherapy for patients with germ cell tumors. METHODS From January 2005 to December 2018, 80 patients were treated with induction chemotherapy for advanced germ cell tumors at Kanagawa Cancer Center Hospital, Japan. Of these, we retrospectively analyzed 267 cycles of chemotherapy. The incidence of FN was used as the objective variable. As predictive factors, we analyzed age, international germ cell consensus classification (IGCCC), laboratory data at the start of chemotherapy in each cycle, length of the largest metastatic lesion, number of cycles, and prophylactic use of granulocyte colony stimulating factor (G-CSF). RESULTS We finally analyzed 267 cycles in 78 patients. The median age was 36 years (15-64). There was a total of 267 cycles. FN occurred in 40 cycles (15%) in 31 patients (40%). The first cycle was accompanied by a significantly higher FN than the subsequent cycles (p < 0.001). The univariate analysis identified age ≧36 years (p = 0.001), creatinine clearance (CCr) <70 (p < 0.001), serum albumin <3.3 (p = 0.002), maximum tumor diameter ≧60 mm (p = 0.036), and first cycle as significant risk factors. The multivariate analysis identified age, CCr, and first cycle as independent predictive factors of FN development. CONCLUSION We identified older age, renal dysfunction, and first cycle of chemotherapy as predictive factors for FN. No statistically significant difference was shown in the usage of prophylactic G-CSF. Special attention should be given to FN in patients with high-risk factors.
Collapse
Affiliation(s)
- Kota Kobayashi
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Takeshi Kishida
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| |
Collapse
|
4
|
Abstract
PURPOSE OF REVIEW The management of testicular cancer has evolved over time with multimodal therapy. Retroperitoneal lymph node dissection (RPLND), which is a complex and potentially morbid treatment option, remains the mainstay in surgical treatment. This article reviews the surgical template, approach and anatomical considerations with regards to nerve spare in RPLND. RECENT FINDINGS The standard full bilateral RPLND template has evolved over time to include the area between the renal hilum, bifurcation of the common iliac vessels, and the ureters. Morbidity with regards to ejaculatory dysfunction has led to further refinements in this procedure. Advancements in anatomical understanding of the retroperitoneal structures and their relationship to the sympathetic chain and hypogastric plexus has allowed for modification of surgical templates. Further refinements in surgical nerve sparing techniques have improved functional outcomes without sacrificing oncological outcomes. Finally, extraperitoneal access to the retroperitoneum and minimally invasive platforms have been implemented to further reduce morbidity. SUMMARY RPLND requires strict adherence to oncological surgical principles regardless of template, approach and technique. Contemporary evidence shows that outcomes are best for advanced testis cancer patients when managed at high volume tertiary care facilities with surgical expertise and access to multidisciplinary care.
Collapse
Affiliation(s)
- Sunny Nalavenkata
- Department of Urology, Westmead Hospital, Westmead, and Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | | | | |
Collapse
|
5
|
Namavari N, Taheri L, Hooshmand F, Dousthaghi MH, Rahmanian V. Pure testicular choriocarcinoma with gastrointestinal metastasis and paraneoplastic symptoms: a case report. BMC Urol 2023; 23:102. [PMID: 37270552 DOI: 10.1186/s12894-023-01271-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 05/09/2023] [Indexed: 06/05/2023] Open
Abstract
BACKGROUND Pure testicular choriocarcinoma is a rare type of non-seminomatous germ cell tumor extremely poor prognostic with the tendency to bleed at the metastatic site. At the time of the diagnosis, 70% of patients have metastatic lesions. Depending on the site of the metastasis, symptoms vary. Gastrointestinal involvement is seen in less than 5% of cases, mostly in the duodenum. CASE PRESENTATION We present a 47 years old male with testicular choriocarcinoma involving the jejunum, lung, liver, and kidney presenting with acute abdominal pain, melena, and dyspnea with some paraneoplastic symptoms. The patient had increased, severe and constant pain in the right lower quadrant for the previous four days. Additionally, he was complaining of nausea, vomiting, anorexia, and a history of melena for the last 10 days. Dyspnea on exertion, hemoptysis, and dry cough were the symptoms he was suffering from, for almost one year. The patient's general appearance was pale, ill, and thin with 10 kg of weight loss during the last some months. The computed tomography (CT) scan reported multiple metastatic lesions in both liver lobes and the left kidney. Pathologic study of the samples of small bowel lesions showed metastatic choriocarcinoma. Following the patient had been referred to an oncologist to start the chemotherapy regime. Finally, the patient has expired after 40 days of his first admission. CONCLUSIONS Testicular choriocarcinoma is a rare but fatal malignancy among young men. Gastrointestinal metastases are infrequent involvement represented by melena and acute abdominal pain, obstruction, and mass. Physicians should consider it as a differential diagnosis for acute abdomen and gastrointestinal bleeding causation.
Collapse
Affiliation(s)
- Negin Namavari
- School of Medicine, Peymaniye Hospital, Jahrom University of Medical Science, Jahrom, Iran.
| | - Lohrasb Taheri
- Department of Surgery, Jahrom University of Medical Science, Jahrom, Iran
| | - Farhang Hooshmand
- Department of pathology, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| | | | - Vahid Rahmanian
- Department of Public Health, Torbat Jam Faculty of Medical Sciences, Torbat Jam, Iran
| |
Collapse
|
6
|
Pisacane A, Cascardi E, Berrino E, Polidori A, Sarotto I, Casorzo L, Panero M, Boccaccio C, Verginelli F, Benvenuti S, Dellino M, Comoglio P, Montemurro F, Geuna E, Marchiò C, Sapino A. Real-world histopathological approach to malignancy of undefined primary origin (MUO) to diagnose cancers of unknown primary (CUPs). Virchows Arch 2023; 482:463-475. [PMID: 36346458 PMCID: PMC9640798 DOI: 10.1007/s00428-022-03435-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/10/2022] [Accepted: 10/15/2022] [Indexed: 11/10/2022]
Abstract
The aim of this study is to envisage a streamlined pathological workup to rule out CUPs in patients presenting with MUOs. Sixty-four MUOs were classified using standard histopathology. Clinical data, immunocytochemical markers, and results of molecular analysis were recorded. MUOs were histologically subdivided in clear-cut carcinomas (40 adenocarcinomas, 11 squamous, and 3 neuroendocrine carcinomas) and unclear-carcinoma features (5 undifferentiated and 5 sarcomatoid tumors). Cytohistology of 7/40 adenocarcinomas suggested an early metastatic cancer per se. In 33/40 adenocarcinomas, CK7/CK20 expression pattern, gender, and metastasis sites influenced tissue-specific marker selection. In 23/40 adenocarcinomas, a "putative-immunophenotype" of tissue of origin addressed clinical-diagnostic examinations, identifying 9 early metastatic cancers. Cell lineage markers were used to confirm squamous and neuroendocrine differentiation. Pan-cytokeratins were used to confirm the epithelial nature of poorly differentiated tumors, followed by tissue and cell lineage markers, which identified one melanoma. In total, 47/64 MUOs (73.4%) were confirmed CUP. Molecular analysis, feasible in 37/47 CUPs (78.7%), had no diagnostic impact. Twenty CUP patients, mainly with squamous carcinomas and adenocarcinomas with putative-gynecologic-immunophenotypes, presented with only lymph node metastases and had longer median time to progression and overall survival (< 0.001), compared with patients with other metastatic patterns. We propose a simplified histology-driven workup which could efficiently rule out CUPs and identify early metastatic cancer.
Collapse
Affiliation(s)
- Alberto Pisacane
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, Turin, Italy
| | - Eliano Cascardi
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, Turin, Italy.
- Department of Medical Sciences, University of Turin, 10100, Turin, Italy.
| | - Enrico Berrino
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, Turin, Italy
- Department of Medical Sciences, University of Turin, 10100, Turin, Italy
| | - Alessio Polidori
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, Turin, Italy
| | - Ivana Sarotto
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, Turin, Italy
| | - Laura Casorzo
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, Turin, Italy
| | - Mara Panero
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, Turin, Italy
| | - Carla Boccaccio
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, Turin, Italy
- Department of Oncology, University of Turin Medical School, 10060, Candiolo, Turin, Italy
| | | | - Silvia Benvenuti
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, Turin, Italy
| | - Miriam Dellino
- Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Paolo Comoglio
- IFOM, FIRC Institute of Molecular Oncology, 20019, Milan, Italy
| | | | - Elena Geuna
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, Turin, Italy
| | - Caterina Marchiò
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, Turin, Italy
- Department of Medical Sciences, University of Turin, 10100, Turin, Italy
| | - Anna Sapino
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, Turin, Italy
- Department of Medical Sciences, University of Turin, 10100, Turin, Italy
| |
Collapse
|
7
|
TIMEAS, a promising method for the stratification of testicular germ cell tumor patients with distinct immune microenvironment, clinical outcome and sensitivity to frontline therapies. Cell Oncol (Dordr) 2023; 46:745-759. [PMID: 36823338 DOI: 10.1007/s13402-023-00781-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2023] [Indexed: 02/25/2023] Open
Abstract
PURPOSE With the heterogeneous genetic background, prognosis prediction and therapeutic targets for testicular germ cell tumors (TGCTs) are still unclear. We defined the tumor immune microenvironment activation status (TIMEAS). METHODS We collected a total of 314 TGCT patients from four cohorts, including a 48-case microarray. A nonnegative matrix factorization algorithm was applied to identify the "immune factor", derived the top 150 weighted genes to divide patients into immune and non-immune classes, and further separated the immune class into activated and exhausted subgroups by nearest template prediction. Tumor mutant burden, gene mutation, and copy number alteration were compared with our recently developed package "MOVICS". A random forest algorithm was performed to establish a prediction model with fewer genes. Immunohistochemistry staining was performed to identify TIMEAS in the microarray. RESULTS We constructed the TIMEAS in the TCGA-TGCT cohort and further validated it in the GSE3218 and GSE99420 cohorts. The immune class contained the activated status of T-lymphocytes, B-lymphocytes, and macrophages, while Treg cells and the WNT/TGFβ signature were more activated in the immune-suppressed subgroup. Patients in the immune-exhausted subgroup had the worst prognosis, and 22.9% of patients in the immune-activated subgroup had KRAS mutations, which might stimulate the response of the immune system and lead to a favorable prognosis. The immune-exhausted group benefited more from chemotherapy, while the immune-activated subgroup responded well to anti-PD-1/PD-L1 therapy. FSCN1 was validated as the target of the immune-exhausted microenvironment by immunohistochemistry. CONCLUSION TIMEAS classification can separate TGCT patients; patients in the immune-activated subgroup could benefit more from anti-PD-L1 immunotherapy, and those in the immune-exhausted subgroup are more suitable for chemotherapy.
Collapse
|
8
|
Vasiliadis K, Simou C, Tzotzou A, Kalinderis N, Valoukas D, Pazarli E, Drakontaidis P, Papavasiliou C. Successful surgical management of massive hemoretroperitoneum caused by spontaneous rupture of retroperitoneal lymph node metastases in a patient with advanced mixed germ cell tumor: a COVID-19 pandemic-related surgical challenge. Surg Case Rep 2023; 9:18. [PMID: 36740640 PMCID: PMC9899871 DOI: 10.1186/s40792-023-01593-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/19/2023] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Spontaneous rapture of a germ cell tumor (GCT) metastases causing massive hemoretroperitoneum in a patient without choriocarcinoma component who has not received previous systemic chemotherapy is an exceedingly rare event. In such a devastating case scenario, a high index of clinical suspicion for early diagnosis and appropriate management is crucial. CASE PRESENTATION We report on a 25-year-old male patient with a 4-month history of orchiectomy for testicular GCT (tGCT), who presented in the emergency department with acute abdomen and hemodynamic instability. Urgent computed tomography scan depicted a retroperitoneal mass measuring approximately 13 × 11.4 × 15 cm and massive intraperitoneal hemorrhage. Hemoperitoneum caused by spontaneous rapture of the metastatic retroperitoneal mass was suspected. COVID-19 pandemic-related deviation from the oncologic surveillance standards combined with COVID-19-related patient's emotional distress and self-neglect had led to loss of opportunity for appropriate adjuvant chemotherapy, obviously leading to the development of this devastating complication. An emergency, surgical exploration was decided. The bleeding mass was adequately exposed following a Cattell-Braasch maneuver and active bleeding was controlled by a challenging resection of approximately 80% of the lymph node mass volume. Pathological evaluation of the specimen revealed teratoma with low volume of yolk sac tumor component and extensive necrosis, findings compatible with the patient's history. Postoperative recovery was uneventful, followed by early start of adjuvant chemotherapy. Two years after the operation the patient is doing well with no evidence of recurrent disease. CONCLUSIONS Massive hemoperitoneum is a devastating event that exceedingly rarely can complicate the clinical course of patients with advanced tGCT. Emergency surgical intervention is usually necessary however, sound judgement and careful surgical techniques are required for a positive and uneventful outcome. During COVID-19 pandemic, first-line medical personnel push their limits further not only to ensure health care services standards but also, to manage unpredictable, life-threatening cancer-related complications, associated with COVID-19-related deviation from appropriate oncologic surveillance and care.
Collapse
Affiliation(s)
- Konstantinos Vasiliadis
- grid.417144.3Surgical Department, Papageorgiou General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Chrysanthi Simou
- grid.417144.3Surgical Department, Papageorgiou General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Anastasios Tzotzou
- grid.417144.3Surgical Department, Papageorgiou General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Kalinderis
- grid.4793.900000001094570052Nd Department of Urology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios Valoukas
- grid.4793.90000000109457005Department of Medical Oncology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Elsa Pazarli
- grid.417144.3Department of Pathology, Papageorgiou General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Paulos Drakontaidis
- grid.417144.3Radiology Department, Papageorgiou General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Christos Papavasiliou
- grid.417144.3Surgical Department, Papageorgiou General Hospital of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
9
|
Terbuch A, Walser G, Stotz M, Gerger A, Posch F, Bauernhofer T. Primary Thromboprophylaxis and the Risk of Venous Thromboembolic Events in Patients With Testicular Germ Cell Tumors Treated With Cisplatinum-Based Chemotherapy. Clin Genitourin Cancer 2023; 21:24-31. [PMID: 36400695 DOI: 10.1016/j.clgc.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/05/2022] [Accepted: 10/09/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND Cisplatinum-based chemotherapy is associated with an increased risk of venous thromboembolism (VTE). We hypothesized that primary thromboprophylaxis in patients with testicular germ cell tumors (GCT) undergoing cisplatinum-based chemotherapy can reduce the risk of VTE. PATIENTS AND METHODS In this single-center retrospective cohort study, we investigated the increased use of primary thromboprophylaxis between January 2000 and December 2021 at our institution and its effect on the risk of VTE. Patients with GCT undergoing adjuvant or curative cisplatinum-based chemotherapy were included. RESULTS Three hundred forty-six patients with GCT initiating a cisplatinum-based therapy were included in the study, of whom 122 (35%) were treated in the adjuvant and 224 (65%) in the curative setting, respectively. VTE events occurred in 49 (14.2%) patients. In univariable competing risk analysis, a higher clinical tumor stage and large retroperitoneal lymphadenopathy (RPLND >5 cm) were the strongest predictors of an elevated VTE risk (SHR for stage IIC - IIIC: 2.6 (95%CI: 5.0-24.7, P < .001), SHR for RPLN: 2.36 (95%CI: 1.27-4.4, P < .007)). The proportion of patients receiving primary thromboprophylaxis strongly increased over time and reached 100% in CS IIC-III patients from 2019 onwards. After adjusting for tumor stage, primary thromboprophylaxis was associated with a 52% relatively lower risk of VTE (SHR = 0.48, 95% CI: 0.24-0.97; P = .032). CONCLUSION In this retrospective cohort study, we showed that TGCT patients undergoing cisplatinum-based chemotherapy have a lower VTE risk when receiving primary thromboprophylaxis. For the duration of chemotherapy, primary thromboprophylaxis should be considered on a risk-benefit ratio.
Collapse
Affiliation(s)
- Angelika Terbuch
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Gudrun Walser
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Michael Stotz
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Armin Gerger
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Florian Posch
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.
| | - Thomas Bauernhofer
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
10
|
Cecilia C, Ardiansyah D, Fadil. A rare case of brain metastatic of primary mediastinal yolk sac tumor. Radiol Case Rep 2023; 18:1041-1045. [PMID: 36684631 PMCID: PMC9849995 DOI: 10.1016/j.radcr.2022.12.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/06/2022] [Accepted: 12/10/2022] [Indexed: 01/07/2023] Open
Abstract
Primary yolk sac tumors are extragonadal germ cell tumors commonly seen in children and young adults. They are more common in men. Germ cells tumor on histopathological characteristics is classified as seminoma and non-seminomatous (NSGC). The rarest form of NSGC is an extragonadal yolk sac tumor of mediastinum. Clinical presentations are not specific and may imitate other chronic disease such as other malignancies or tuberculosis such as chest discomfort, vena cava superior syndrome, fever, weight loss, and chronic cough. Immunohistochemistry showed a positive result in Alpha-fetoprotein and pan-cytokeratin. Due to its rarity, brain metastases' clinical signs and symptoms, anatomical sites, and characteristics are less well documented. However, the metastatic brain process gave similar histological findings to the primary site. Additional radiological and laboratory tests can be carried out to identify other metastatic processes. Standardized treatment of primary mediastinal sac tumors with brain metastasis has not yet been established. Combining chemotherapy, surgery and radiation treatment could improve overall outcomes and prognosis. We present a scarce case of primary mediastinal yolk sac tumor with metastatic brain process in a 32-year-old male with a short survival period.
Collapse
|
11
|
Filho LHCP, Silva TA, Beal PR, Buonfiglio VB, Sadi MV. Spontaneous rupture of a retroperitoneal lymph node metastasis causing massive hemorrhage in a patient with advanced mixed germ cell tumor. Urol Case Rep 2022; 45:102180. [PMID: 35968525 PMCID: PMC9372596 DOI: 10.1016/j.eucr.2022.102180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/28/2022] [Accepted: 07/31/2022] [Indexed: 11/30/2022] Open
Abstract
Retroperitoneal hemorrhage is a rare and life-threatening clinical presentation that may occur in patients with advanced germ cell tumors, typically after chemotherapy. Rapid clinical suspicion is crucial to offer the best treatment. Surgery is usually recommended but dismal complications may occur. We report on a man with metastatic testicular cancer presenting with massive spontaneous retroperitoneal hemorrhage that had not received previous systemic treatment and was managed without surgical treatment, with good clinical outcome.
Collapse
Affiliation(s)
| | - Tiago Aparecido Silva
- Corresponding author. Department of Surgery, Division of Urology Federal University of São Paulo Rua Botucatu, 740 – Vila Clementino, Zip Code 04023062, São Paulo, SP, Brazil.
| | | | | | | |
Collapse
|
12
|
Lesko P, Chovanec M, Mego M. Biomarkers of disease recurrence in stage I testicular germ cell tumours. Nat Rev Urol 2022; 19:637-658. [PMID: 36028719 DOI: 10.1038/s41585-022-00624-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2022] [Indexed: 11/09/2022]
Abstract
Stage I testicular cancer is a disease restricted to the testicle. After orchiectomy, patients are considered to be without disease; however, the tumour is prone to relapse in ~4-50% of patients. Current predictive markers of relapse, which are tumour size and invasion to rete testis (in seminoma) or lymphovascular invasion (in non-seminoma), have limited clinical utility and are unable to correctly predict relapse in a substantial proportion of patients. Adjuvant therapeutic strategies based on available biomarkers can lead to overtreatment of 50-85% of patients. Discovery and implementation of novel biomarkers into treatment decision making will help to reduce the burden of adjuvant treatments and improve patient selection for adjuvant therapy.
Collapse
Affiliation(s)
- Peter Lesko
- 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Michal Chovanec
- 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.
| |
Collapse
|
13
|
Wang S, Yang X, Yu Z, Du P, Cao Y, Ji Y, Ma J, Yang Y. The values of systemic immune-inflammation index and neutrophil-lymphocyte ratio in predicting testicular germ cell tumors: A retrospective clinical study. Front Oncol 2022; 12:893877. [PMID: 36185298 PMCID: PMC9523471 DOI: 10.3389/fonc.2022.893877] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 09/01/2022] [Indexed: 11/26/2022] Open
Abstract
Purpose To determine whether complete blood count (CBC) based inflammatory parameters can be used as markers predicting testicular germ cell tumors (TGCT). Material and methods Between 2013 to 2018 the data of 58 patients with testicular TGCT undergoing radical orchiectomy and 54 malignancy-free healthy men were retrospectively analyzed as tumor group and control group. Patient baseline characteristics including age, pathological stage and pre-surgery CBC based inflammatory parameters including neutrophil/lymphocyte ratio (NLR), platelet/lymphocyte ratio (PLR), lymphocyte/monocyte ratio (LMR), systemic immune-inflammation index (SII), lymphocyte ratio (LR), neutrophil ratio (NR), mean platelet volume (MPV) and red cell distribution width (RDW) were analyzed and compared between tumor group and control group. Receiver operating characteristic (ROC) curve were used analyzing data with significantly difference to assess the discriminative ability of the markers for TGCT, area under the curve (AUC), cut-off value, sensitivity and specificity were calculated. The binary logistic regression model was used to evaluate the association between significant inflammatory markers and risk of TGCT. Results Mean age of the tumor and control group was 41.1 ± 15.36 and 44.89 ± 9.2 years, respectively. Mean NLR, SII and RDW were significantly higher in tumor group compared with control group with P=0.005, P=0.001 and P=0.016, respectively; there were no significantly differences of age, PLR, LMR, LR, NR, MPV and RDW between groups. The ROC curve for NLR, SII and RDW was plotted in the diagnosis of TGCT and tumor progression, the cut-off value for NLR, SII and RDW were found as 3.38 (AUC: 0.704, sensitivity=51.4%, specificity=88.6%, P=0.003), 881.24 (AUC: 0.725, sensitivity=45.7%, specificity=91.4%, P=0.001) and 0.14 (AUC: 0.63, sensitivity=28.6%, specificity=97%, P=0.063), respectively. Patients were divided into two groups according to the threshold values, respectively. By using the multivariable logistic regression models, NLR ≥ 3.38 (OR, 5.86; 95% CI, 1.67-20.65, P=0.006) and SII ≥ 881.24 (OR, 4.89; 95% CI, 1.48-15.32, P=0.009) were independent risk factors predicting TGCT. Significantly statistical difference of pathological stage was also found between groups with respect to NLR cut-off values (P=0.034) and SII cut-off values (P=0.049). Combined the data together, NLR and SII both exhibited good differential diagnosis potential which could be used as markers predicting the TGCT. Conclusion As the CBC based inflammation parameters, both NLR and SII could be used as effective tumor markers predicting the TGCT, and higher NLR and SII are associated with higher pathological stage. In addition, SII is a more powerful tool among these two inflammatory markers.
Collapse
|
14
|
Yumioka T, Morizane S, Makishima K, Adachi T, Moriyasu E, Iwamoto H, Hikita K, Honda M, Umekita Y, Takenaka A. Brain and spinal cord metastases with seminoma: A case report. IJU Case Rep 2022; 5:464-468. [PMID: 36341180 PMCID: PMC9626331 DOI: 10.1002/iju5.12515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/20/2022] [Indexed: 12/03/2022] Open
Abstract
Introduction Brain and spinal cord metastases from testicular cancer occur rarely, and metastases with seminoma are extremely rare. Case presentation A 42‐year‐old man who was diagnosed with seminoma and multiple metastases underwent first‐line and salvage chemotherapy. Brain metastases were noted; consequently, surgery, third‐line chemotherapy, and whole‐brain irradiation were performed. Subsequently, paralysis developed, and spinal cord metastases were detected. He received fourth‐line chemotherapy but died. Pathological autopsy revealed metastases only in the spinal cord. The cause of death was considered respiratory failure due to cervical spinal cord involvement from spinal metastases. Conclusion Brain and spinal cord metastases from seminoma are rare. Thus, similar future cases should be treated appropriately.
Collapse
Affiliation(s)
- Tetsuya Yumioka
- Division of Urology, Department of Surgery, Faculty of Medicine Tottori University Tottori Japan
| | - Shuichi Morizane
- Division of Urology, Department of Surgery, Faculty of Medicine Tottori University Tottori Japan
| | - Karen Makishima
- Division of Pathology, Department of Pathology, Faculty of Medicine Tottori University Tottori Japan
| | - Tadashi Adachi
- Division of Neuropathology, Department of Brain and Neurosciences, Faculty of Medicine Tottori University Tottori Japan
| | - Emika Moriyasu
- Division of Urology, Department of Surgery, Faculty of Medicine Tottori University Tottori Japan
| | - Hideto Iwamoto
- Division of Urology, Department of Surgery, Faculty of Medicine Tottori University Tottori Japan
| | - Katsuya Hikita
- Division of Urology, Department of Surgery, Faculty of Medicine Tottori University Tottori Japan
| | - Masashi Honda
- Division of Urology, Department of Surgery, Faculty of Medicine Tottori University Tottori Japan
| | - Yoshihisa Umekita
- Division of Pathology, Department of Pathology, Faculty of Medicine Tottori University Tottori Japan
| | - Atsushi Takenaka
- Division of Urology, Department of Surgery, Faculty of Medicine Tottori University Tottori Japan
| |
Collapse
|
15
|
Qin L, Wang B, Wang Z, He D. Clinicopathological features, prognosis, and fertility outcomes in Chinese Han women treated for ovarian yolk sac tumor: A retrospective case series study from two tertiary-care academic medical centers. Medicine (Baltimore) 2022; 101:e29868. [PMID: 35866762 PMCID: PMC9302259 DOI: 10.1097/md.0000000000029868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE Ovarian yolk sac tumor (YST) is a very rare malignant tumor in young women. This study aimed to explore the clinicopathological prognostic characteristics and reproductive outcomes of Chinese Han patients. METHODS To describe a case series study, we reviewed the clinicopathological data of 50 YST patients treated from 2 tertiary medical academic medical centers from January 2009 to December 2019. The Akaike information criterion was used to select variables. The influence of relevant characteristics on prognosis factors was analyzed by the Cox proportional hazard model. RESULTS The median follow-up time was 64.5 months (range from 3 to 124 months). The median age was 22.7 years (3 to 34 years). Abdominal pain (54.0%) or mass (42.0%) were the most common clinical symptoms in the early stage of diagnosis. The tumors were located bilaterally in 4 cases. 27 patients, 7 patients, 13 patients, and 3 patients were in stage I, II, III, and IV, respectively. Twenty-one stage I patients and 12 stage II to IV patients underwent fertility-preserving surgery. Of the 50 patients who received postoperative chemotherapy, 49 received the BEP regimen. At the last follow-up, 92% of the patients were still alive. The overall survival rate and disease-free survival rate were 91.6% and 90.6%, respectively. Recurrence occurred in 7 (14%) patients with a median survival time of 16.7 months (range from 3 to 50 months). Six patients had recurrence in the abdominal space. The percentage of Ki67 (P = .01) and tumor size (P = .03) were 2 important prognostic factors in multivariate analysis. In terms of survival outcomes, fertility-preserving surgery can be equivalent to radical surgery. Sixteen patients tried to conceive, and 6 patients with advanced-stage succeeded in 10 pregnancies. Of these, 6 patients successfully gave birth to 6 healthy babies. CONCLUSIONS The diagnosis of YST of childbearing age is very rare. Because the failure of primary treatment is related to the residual disease after salvage surgery, the fertility and survival results of patients undergoing fertility-preserving surgery are promising.
Collapse
Affiliation(s)
- Li Qin
- Department of Obstetrics and Gynecology, The Central Hospital of Enshi Tujia and Miao Autonomous prefecture, Enshi, Hubei, China
| | - Bo Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zaiping Wang
- Department of Obstetrics and Gynecology, The Central Hospital of Enshi Tujia and Miao Autonomous prefecture, Enshi, Hubei, China
| | - Du He
- Department of Obstetrics and Gynecology, The Central Hospital of Enshi Tujia and Miao Autonomous prefecture, Enshi, Hubei, China
| |
Collapse
|
16
|
Pini GM, Colecchia M. Mediastinal germ cell tumors: a narrative review of their traits and aggressiveness features. MEDIASTINUM (HONG KONG, CHINA) 2022; 6:5. [PMID: 35340833 PMCID: PMC8841550 DOI: 10.21037/med-21-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/06/2021] [Indexed: 05/25/2023]
Abstract
OBJECTIVE Mediastinal extragonadal germ-cell tumors (MEGCTs) are rare neoplasms with a multifaceted clinical behavior. This paper is devoted to review their main characteristics, including histological patterns and different factors of aggressiveness in MEGCTs. Proper understanding of the latter can help to better stratify patients' prognoses and improve clinical management. BACKGROUND Different theories exist on the origin of MEGCTs, including primordial germ cells deposition during embryogenesis. MEGCTs predominantly affects young males and aggressiveness follows the ability of local and systemic spread of each germ-cell neoplasia subtype, as well as their distinct responsiveness to therapy. Indeed, non-seminomatous MEGCTs have a worse prognosis. Unfortunately, they are also more frequent than seminomas in the mediastinum. Regardless of histological type, local aggressiveness can follow tumoral expansion with compression on or infiltration of mediastinal structures. Chemotherapy can be effective in reducing neoplastic volume, but different levels of sensitivity can be found in different MGCTs. In particular, a chemo-resistant teratoma component of a mixed MEGCTs can undergo a paradoxical enlargement after chemotherapy, while other components of the tumor regress. This is reflected by a concomitant normalization of serum tumoral markers and cardiopulmonary deterioration due to compression. Such clinical phenomenon, called growing-teratoma syndrome (GTS), requires a prompt surgical approach. METHODS A literature research of pertinent epidemiological, pathological and clinical articles was conducted. CONCLUSION The mediastinum can harbor different kinds of neoplasia, including GCTs. The full spectrum of MEGCTs includes a variety of tumors with different clinical behaviors. Aggressiveness follows the inherent ability of local and systemic spread of each neoplastic type, as well as their distinct responsiveness to therapy.
Collapse
Affiliation(s)
- Giacomo M. Pini
- Department of Medicine and Surgery, Unit of Pathology, University of Insubria, Varese, Italy
| | - Maurizio Colecchia
- Department of Pathology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
17
|
Storey B, Raman A, Grant A. Metachronous testicular tumour recurrence in extra-gonadal germ cell tumour: A case report and review of the literature. JOURNAL OF CLINICAL UROLOGY 2021. [DOI: 10.1177/2051415819892763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A 24-year-old male presented in 2014 with left sided abdominal pain and a palpable abdominal mass. After appropriate investigation a diagnosis of an extra-gonadal germ cell tumour was made and he was treated with platinum based chemotherapy. He then underwent retroperitoneal lymph node dissection due to residual disease identified on imaging with no specific tissue type identified on the histological sample. In early 2019 he presented to his General Practitioner with a left sided testicular lump and was diagnosed with non-seminomatous germ cell tumour of the testis, an uncommon condition; metachronous testicular tumour. Level of Evidence: 5
Collapse
Affiliation(s)
- Benjamin Storey
- Department of Urology, Royal Newcastle Centre, New Lambton Heights, Australia
| | - Avi Raman
- Department of Urology, Royal Newcastle Centre, New Lambton Heights, Australia
| | - Alexander Grant
- Department of Urology, Royal Newcastle Centre, New Lambton Heights, Australia
| |
Collapse
|
18
|
Zhu J, Chen H, Chen T, Zhen Z, Wang J, Sun F, Lu S, Huang J, Zhang Y, Sun X. Multimodal Treatment of Children With Sacrococcygeal Yolk Sac Tumor: Retrospective Analysis of Clinicopathology Characteristics and Relapse-free Survival. J Pediatr Hematol Oncol 2021; 43:e848-e853. [PMID: 33512871 PMCID: PMC8327933 DOI: 10.1097/mph.0000000000002068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 12/06/2020] [Indexed: 11/19/2022]
Abstract
The aim of the study was to explore the clinicopathologic characteristics of sacrococcygeal yolk sac tumor (SYST) associated with relapse and the role of sensitivity to neoadjuvant chemotherapy in predicting outcome. The authors investigated prognostic factors of age, stage, initial tumor size, pathologic response to neoadjuvant chemotherapy, and alfa fetoprotein. A total of 26 patients with SYST were enrolled. Neoadjuvant chemotherapy was administered to 20 cases. Six patients underwent resection as initial therapy. Recurrence occurred in 12 patients. Nine patients with specimens exhibiting no malignant component after chemotherapy did not experience recurrence. By contrast, relapses occurred in 7 of 11 patients with viable residual tumor after neoadjuvant chemotherapy. All relapsed patients still achieved partial remission or complete remission after salvage therapy. Five-year relapse-free survival and overall survival rates were 55.2% and 100%, respectively (median follow-up, 59.5 mo; range, 16 to 155). Patients with complete necrosis after neoadjuvant chemotherapy had a better outcome compared with children with viable residual tumor. Relapse-free survival of pediatric SYSTs in this cohort were still low and warrants the multidisciplinary effort.
Collapse
Affiliation(s)
- Jia Zhu
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center
- Key Laboratory of Oncology in South China
- Collaborative Innovation Center for Cancer Medicine
| | - Huadong Chen
- Department of Pediatric Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Zhongshan Er Lu, Guangzhou, China
| | - Tingting Chen
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center
- Key Laboratory of Oncology in South China
- Collaborative Innovation Center for Cancer Medicine
| | - Zijun Zhen
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center
- Key Laboratory of Oncology in South China
- Collaborative Innovation Center for Cancer Medicine
| | - Juan Wang
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center
- Key Laboratory of Oncology in South China
- Collaborative Innovation Center for Cancer Medicine
| | - Feifei Sun
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center
- Key Laboratory of Oncology in South China
- Collaborative Innovation Center for Cancer Medicine
| | - Suying Lu
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center
- Key Laboratory of Oncology in South China
- Collaborative Innovation Center for Cancer Medicine
| | - Junting Huang
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center
- Key Laboratory of Oncology in South China
- Collaborative Innovation Center for Cancer Medicine
| | - Yizhuo Zhang
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center
- Key Laboratory of Oncology in South China
- Collaborative Innovation Center for Cancer Medicine
| | - Xiaofei Sun
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center
- Key Laboratory of Oncology in South China
- Collaborative Innovation Center for Cancer Medicine
| |
Collapse
|
19
|
Trends in the use of administrative databases in urologic oncology: 2000–2019. Urol Oncol 2021; 39:487-492. [DOI: 10.1016/j.urolonc.2021.01.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 12/11/2022]
|
20
|
Darif K, Benbrahim Z, Acharfi N, Khacha A, Maaroufi M, Amaadour L, Oualla K, Arifi S, Mellas N. A primary mediastinal germ cell tumor of yolk sac type: case report. Pan Afr Med J 2021; 38:330. [PMID: 34285753 PMCID: PMC8265249 DOI: 10.11604/pamj.2021.38.330.23730] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 03/16/2021] [Indexed: 11/11/2022] Open
Abstract
The mediastinal malignant germ cells tumor represents less than 0.5% of thoracic tumors, although the mediastinum is one of the main extragonadic locations of these tumors. In the majority of cases, young people are those most affected. The prognosis of mediastinal malignant germ cells tumors is poor, especially non-seminomatous germ tumors. In this article, we report a rare case of a young 19-years-old patient treated for a mediastinal germ cell tumor of yolk sac. The patient presented a chest pain; the chest computed tomography (CT) showed a right paramedian mediastinal mass with a pleural effusion associated with supraclavicular and cervical lymph nodes. Biopsy revealed a non-seminomatousgerm cell tumor of yolk sac. The exams showed elevated alpha-fetoprotein (AFP), without any meaningful elevation of other serictumor markers. The patient received 4 cycles of chemotherapy based on etoposide, ifosfamide and platinum salts then a complete excision of the mass.
Collapse
Affiliation(s)
- Khadija Darif
- Department of Medical Oncology, Hassan II University Hospital, Fez, Morocco.,Faculty of Medicine and Pharmacy of Fez, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Zineb Benbrahim
- Department of Medical Oncology, Hassan II University Hospital, Fez, Morocco.,Faculty of Medicine and Pharmacy of Fez, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Nisrine Acharfi
- Department of Medical Oncology, Hassan II University Hospital, Fez, Morocco.,Faculty of Medicine and Pharmacy of Fez, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Anass Khacha
- Faculty of Medicine and Pharmacy of Fez, Sidi Mohamed Ben Abdellah University, Fez, Morocco.,Department of Radiology, Hassan II University Hospital, Fez, Morocco
| | - Mustapha Maaroufi
- Faculty of Medicine and Pharmacy of Fez, Sidi Mohamed Ben Abdellah University, Fez, Morocco.,Department of Radiology, Hassan II University Hospital, Fez, Morocco
| | - Lamiae Amaadour
- Department of Medical Oncology, Hassan II University Hospital, Fez, Morocco.,Faculty of Medicine and Pharmacy of Fez, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Karima Oualla
- Department of Medical Oncology, Hassan II University Hospital, Fez, Morocco.,Faculty of Medicine and Pharmacy of Fez, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Samia Arifi
- Department of Medical Oncology, Hassan II University Hospital, Fez, Morocco.,Faculty of Medicine and Pharmacy of Fez, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Nawfel Mellas
- Department of Medical Oncology, Hassan II University Hospital, Fez, Morocco.,Faculty of Medicine and Pharmacy of Fez, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
21
|
Guo H, Chen H, Wang W, Chen L. Clinicopathological Features, Prognostic Factors, Survival Trends, and Treatment of Malignant Ovarian Germ Cell Tumors: A SEER Database Analysis. Oncol Res Treat 2021; 44:145-153. [PMID: 33706324 DOI: 10.1159/000509189] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/05/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the clinicopathological prognostic factors of malignant ovarian germ cell tumors (MOGCT) and evaluate the survival trends of MOGCT by histotype. METHODS We extracted data on 1,963 MOGCT cases diagnosed between 2000 and 2014 from the Surveillance, Epidemiology, and End Results (SEER) database and the histological classification of MOGCT, including 5 categories: dysgerminoma, embryonal carcinoma (EC), yolk sac tumor, malignant teratoma, and mixed germ cell tumor. We examined overall and disease-specific survival of the 5 histological types. Kaplan-Meier and Cox proportional hazards regression models were used to estimate survival curves and prognostic factors. We also estimated survival curves of MOGCT according to different treatments. RESULTS There was a significant difference in prognosis among different histological classifications. Age, histotype, grade, SEER stage, and surgery were independent prognostic factors for survival of patients with MOGCT. For all histotypes, 1-, 3-, and 5-year survival rate estimates were >85%, except for EC, which had the worst outcomes at 1 year (55.6%), 3 years (44.4%), and 5 years (33.3%). In the distant SEER stage, both chemotherapy and surgery were associated with improved survival outcomes compared with surgery- and chemotherapy-only groups. CONCLUSIONS Dysgerminoma patients had the most favorable outcomes, whereas EC patients had the worst survival. A young age, low grade, and surgery were all significant predictors for improved survival. In contrast, a distant SEER stage was a risk factor for poor survival. Chemotherapy combined with surgery contributed to longer survival times of patients with MOGCT in the distant SEER stage.
Collapse
Affiliation(s)
- Hualei Guo
- Department of Pathology, Hangzhou Women's Hospital, Hangzhou, China,
| | - Hao Chen
- Department of Pathology, Hangzhou Women's Hospital, Hangzhou, China
| | - Wenhui Wang
- Department of Pathology, Hangzhou Women's Hospital, Hangzhou, China
| | - Lingna Chen
- Department of Gynecology, Hangzhou Women's Hospital, Hangzhou, China
| |
Collapse
|
22
|
Gastrointestinal Bleeding Secondary to Metastatic Duodenal Choriocarcinoma in a Patient with Concomitant Peptic Ulcer Disease. Case Rep Gastrointest Med 2021; 2021:6664147. [PMID: 33763267 PMCID: PMC7946466 DOI: 10.1155/2021/6664147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/23/2021] [Indexed: 11/23/2022] Open
Abstract
Testicular tumors are one of the most common solid tumors in young males. Choriocarcinoma usually presents as metastatic disease. Gastrointestinal tract involvement is rare. We report a case of a 40-year-old male presenting to our hospital with a three-day history of dyspnea on exertion and black stool after recent diagnosis of testicular choriocarcinoma. Urgent EGD performed revealed small clean-based fundal ulcer and an antral ulcer without the stigma of recent bleeding. Capsule endoscopy was performed and revealed a bleeding ill-defined mass in the proximal duodenum. A subsequent push enteroscopy showed an ulcerated bleeding mass in the third part of the duodenum that was treated with a hemospray with adequate hemostasis. Pathology was consistent with pure choriocarcinoma. The patient received a cisplatin-based chemotherapy regimen. The patient tolerated the chemotherapy regimen well and was discharged for outpatient follow-up. At the three-month follow-up, the patient did not show evidence of recurrent gastrointestinal bleeding.
Collapse
|
23
|
Revels JW, Wang SS, Gangadhar K, Ali A, Ali AA, Lee JH. Multimodality Radiological Pictorial Review of Testicular Carcinoma: From Initial Staging to Restaging. Res Rep Urol 2020; 12:599-613. [PMID: 33294422 PMCID: PMC7718994 DOI: 10.2147/rru.s257243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/27/2020] [Indexed: 11/23/2022] Open
Abstract
With an overall 5-year survival rate >95%, patients with testicular cancer have a great prognosis. Although initial diagnosis is based on clinical examination, imaging does play a significant role in the diagnosis and prognosis of testicular cancer, which are dependent on tumor burden and staging. Successful treatment requires appropriate disease assessment throughout a patient’s treatment: evaluating treatment response, restaging, and monitoring for disease recurrence after treatment completion. Ultrasound is usually the initial screening modality for painless testicular masses, and computedtomography (CT) the most commonly used for staging and restaging. However, with regard to seminomas, positron-emission tomography (PET) combined with CT is slowly taking priority. With regard to nonseminomatous germ-cell tumors, PET-CT has not proven to be completely effective, due to a high number of false-negative results. The purpose of this paper is to provide radiologists with a pictorial review of testicular carcinoma from initial staging through posttreatment follow-up.
Collapse
Affiliation(s)
- Jonathan W Revels
- Department of Radiology, University of New Mexico, Albuquerque, NM, USA
| | - Sherry S Wang
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake, UT, USA
| | - Kiran Gangadhar
- Department of Radiology, University of Washington Medical Center, Seattle, WA, USA
| | - Arafat Ali
- Department of Radiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Al-Amin Ali
- Department of Literature, Science, and Arts, University of Michigan, Ann Arbor, MI, USA
| | - Jean H Lee
- Department of Radiology, University of Washington Medical Center, Seattle, WA, USA
| |
Collapse
|
24
|
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
|
25
|
Groot HJ, van Leeuwen FE, Lubberts S, Horenblas S, de Wit R, Witjes JA, Groenewegen G, Poortmans PM, Hulshof MCCM, Meijer OWM, de Jong IJ, van den Berg HA, Smilde TJ, Vanneste BGL, Aarts MJB, Jóźwiak K, van den Belt-Dusebout AW, Gietema JA, Schaapveld M. Platinum exposure and cause-specific mortality among patients with testicular cancer. Cancer 2019; 126:628-639. [PMID: 31730712 PMCID: PMC7004069 DOI: 10.1002/cncr.32538] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 11/06/2022]
Abstract
Background Although testicular cancer (TC) treatment has been associated with severe late morbidities, including second malignant neoplasms (SMNs) and ischemic heart disease (IHD), cause‐specific excess mortality has been rarely studied among patients treated in the platinum era. Methods In a large, multicenter cohort including 6042 patients with TC treated between 1976 and 2006, cause‐specific mortality was compared with general population mortality rates. Associations with treatment were assessed with proportional hazards analysis. Results With a median follow‐up of 17.6 years, 800 patients died; 40.3% of these patients died because of TC. The cumulative mortality was 9.6% (95% confidence interval [CI], 8.5%‐10.7%) 25 years after TC treatment. In comparison with general population mortality rates, patients with nonseminoma experienced 2.0 to 11.6 times elevated mortality from lung, stomach, pancreatic, rectal, and kidney cancers, soft‐tissue sarcomas, and leukemia; 1.9‐fold increased mortality (95% CI, 1.3‐2.8) from IHD; and 3.9‐fold increased mortality (95% CI, 1.5‐8.4) from pneumonia. Seminoma patients experienced 2.5 to 4.6 times increased mortality from stomach, pancreatic, bladder cancer and leukemia. Radiotherapy and chemotherapy were associated with 2.1 (95% CI, 1.8‐2.5) and 2.5 times higher SMN mortality (95% CI, 2.0‐3.1), respectively, in comparison with the general population. In a multivariable analysis, patients treated with platinum‐containing chemotherapy had a 2.5‐fold increased hazard ratio (HR; 95% CI, 1.8‐3.5) for SMN mortality in comparison with patients without platinum‐containing chemotherapy. The HR for SMN mortality increased 0.29 (95% CI, 0.19‐0.39) per 100 mg/m2 platinum dose administered (Ptrend < .001). IHD mortality was increased 2.1‐fold (95% CI, 1.5‐4.2) after platinum‐containing chemotherapy in comparison with patients without platinum exposure. Conclusions Platinum‐containing chemotherapy is associated with a dose‐dependent increase in the risk of SMN mortality. Platinum‐containing chemotherapy is associated with a dose‐dependent increase in the risk of cancer mortality among patients with testicular cancer. Patients with testicular cancer experience increased mortality from second malignancies as well as causes other than cancer, particularly ischemic heart diseases.
Collapse
Affiliation(s)
- Harmke J Groot
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Flora E van Leeuwen
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sjoukje Lubberts
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Simon Horenblas
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ronald de Wit
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - J Alfred Witjes
- Department of Urology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Gerard Groenewegen
- Department of Medical Oncology, Cancer Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Philip M Poortmans
- Department of Radiation Oncology, Dr. Bernard Verbeeten Institute, Tilburg, the Netherlands.,Department of Radiation Oncology, Curie Institute, Paris, France
| | - Maarten C C M Hulshof
- Department of Radiation Oncology, Academic Medical Center, Amsterdam, the Netherlands
| | - Otto W M Meijer
- Department of Radiation Oncology, VU University Medical Center Amsterdam, Amsterdam, the Netherlands
| | - Igle J de Jong
- Department of Urology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Tineke J Smilde
- Department of Medical Oncology, Jeroen Bosch Hospital, 's-Hertogenbosch, the Netherlands
| | - Ben G L Vanneste
- Department of Radiotherapy, Maastro Clinic, Maastricht, the Netherlands
| | - Maureen J B Aarts
- Department of Medical Oncology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Katarzyna Jóźwiak
- Department of Biostatistics, Netherlands Cancer Institute, Amsterdam, the Netherlands.,Institute of Biostatistics and Registry Research, Brandenburg Medical School-Theodor Fontane, Neuruppin, Germany
| | | | - Jourik A Gietema
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Michael Schaapveld
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
26
|
Kosti K, Athanasiadis L, Goulis DG. Long-term consequences of androgen insensitivity syndrome. Maturitas 2019; 127:51-54. [PMID: 31351520 DOI: 10.1016/j.maturitas.2019.06.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 06/02/2019] [Accepted: 06/04/2019] [Indexed: 11/18/2022]
Abstract
Androgen insensitivity syndrome (AIS) is one of the most common sexual developmental disorders. According to the grade of the remaining androgen receptor (AR) function, AIS is classified as complete (CAIS), partial (PAIS) or mild (MAIS). In CAIS, the prevalence of germ cell tumours is increased compared with the general population. Although patients with CAIS used to undergo gonadectomy before puberty, nowadays a gonadectomy is recommended after spontaneous puberty, and up to 15% of patients retain their gonads. Nevertheless, the risk of germ cell tumour increases gradually after puberty. Annual follow-up with ultrasound or magnetic resonance imaging (MRI) is recommended. Unfortunately, these imaging methods are not sensitive enough for the diagnosis of an in situ germ cell tumour. In PAIS, the risk of germ cell tumour is higher than in CAIS; therefore, an early gonadectomy or an orchidopexy is indicated. Optimal hormone replacement therapy (HRT) is necessary for long-term health. The risks of osteopenia and of regimen osteoporosis are higher, ESPECIALLY in patients with early gonadectomy. Infertility is the rule in CAIS and PAIS. A few mutations do not affect fertility detrimentally, and these are responsible for MAIS. In PAIS leading to a predominantly male phenotype or ambiguous genitalia, multiple surgical procedures for gynaecomastia and/or hypospadias are required. Some small studies have found a higher risk of obesity, hyperlipidaemia and impaired insulin sensitivity. Psychological support is essential, as the prevalence of psychiatric disorders is increased. In conclusion, the diagnosis of AIS has long-term consequences for which shared decision-making (physicians, patients, parents) is appropriate.
Collapse
Affiliation(s)
- Konstantia Kosti
- Unit of Reproductive Endocrinology, 1(st) Department of Obstetrics and Gynaecology, Medical School, Aristotle University of Thessaloniki, Greece.
| | - Loukas Athanasiadis
- Third Department of Psychiatry, Medical School, Aristotle University of Thessaloniki, Greece
| | - Dimitrios G Goulis
- Unit of Reproductive Endocrinology, 1(st) Department of Obstetrics and Gynaecology, Medical School, Aristotle University of Thessaloniki, Greece
| |
Collapse
|
27
|
Abstract
Experience demonstrates multiple paths to cure for patients with clinical stage I testicular cancer. Because all options should provide a long-term disease-free rate near 100%, overall survival is no longer relevant in decision making, allowing practitioners to factor in quality of life, toxicity, cost, and impact on compliance. Surveillance for clinical stage I seminoma and clinical stage I nonseminoma has become the preferred option. The contrarian view is that a risk-adapted approach should persist, with surveillance for low-risk individuals and active therapy high-risk individuals. However, results obtained in unselected patients provide a strong argument against the need for such an approach.
Collapse
Affiliation(s)
- Bruce J Roth
- Division of Oncology/BMT, Department of Medicine, Washington University in St. Louis, 660 South Euclid Avenue, CB 8056, St Louis, MO 63110, USA.
| |
Collapse
|
28
|
Purnamasidhi CAW, Suega K, Bakta IM. Association between Lactate Dehydrogenase Levels to the Response of Non-Hodgkin Lymphoma in Elderly Patients Who Treated with First-Line Chemotherapy in Sanglah General Hospital. Open Access Maced J Med Sci 2019; 7:1984-1986. [PMID: 31406541 PMCID: PMC6684432 DOI: 10.3889/oamjms.2019.470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND: Non-Hodgkin Lymphoma (NHL) is a malignant haematological disease originates in the lymphocytes, caused by an abnormality in lymphocytes development which forms a tumour and may become cancer. Chemotherapy is the main treatment modality for aggressive lymphoma, but only a few patients achieve remission. Several factors such as age, clinical stadium, number of extranodal regions, and Lactate Dehydrogenase (LDH) level played a role in determining response to chemotherapy. AIM: To measure the association between LDH levels to prognosis of NHL in elderly patients who treated with rituximab, cyclophosphamide, doxorubicin, vincristine and prednisolone chemotherapy in Sanglah General Hospital. METHODS: This study used a retrospective descriptive study on elderly NHL patients in Sanglah General Hospital from January until December 2014. The evaluation was measured using the IPI score to determine the prognosis of patients. Demographic data, the stadium, extranodal region, LDH level, and response to chemotherapy were recorded. RESULTS: Twenty-five patients were included in the study. The age ranged was between 61-76 years old (Mean 65,68 ± 4,7 years; Median 65 years). The number of male patients was 19 (76%). Diffuse Large B-Cell Lymphoma (DLBCL) is the most common histopathological structure observed on the patients (68%). LDH levels were normal in 51.6% of the patients and considered high in the rests (48.4%). Results of the chemotherapy were a good response in 72.2%. Compared to the patients who showed complete response to chemotherapy, patients with no response (partial response and progression) had significantly higher levels of LDH (OR: 13,1; 95% CI: 1,36-126,30; p = 0,001). CONCLUSION: Non-Hodgkin Lymphoma in elderly patients with no response to chemotherapy had significantly higher levels of LDH than patients with complete response.
Collapse
Affiliation(s)
- Cokorda Agung Wahyu Purnamasidhi
- Division of Hematology and Medical Oncology, Internal Medicine Department Udayana University, Sanglah General Hospital Denpasar, Denpasar, Bali, Indonesia
| | - Ketut Suega
- Division of Hematology and Medical Oncology, Internal Medicine Department Udayana University, Sanglah General Hospital Denpasar, Denpasar, Bali, Indonesia
| | - I Made Bakta
- Division of Hematology and Medical Oncology, Internal Medicine Department Udayana University, Sanglah General Hospital Denpasar, Denpasar, Bali, Indonesia
| |
Collapse
|
29
|
Lehnich AT, Rusner C, Chodick G, Katz R, Sella T, Stang A. Actual frequency of imaging during follow-up of testicular cancer in Israel-a comparison with the guidelines. Eur Radiol 2019; 29:3918-3926. [PMID: 31016446 DOI: 10.1007/s00330-019-06148-1] [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: 10/15/2018] [Revised: 02/01/2019] [Accepted: 03/08/2019] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Computed tomography (CT) examinations are frequent in follow-up care of testicular cancer (TC) but may increase the risk for other cancers. We wanted to assess the actual number of CT and X-ray examinations within the first 5 years after a diagnosis of TC in Israel during 2003-2007. METHODS The database of Maccabi Healthcare Services, Israel, was searched for TC patients diagnosed in 2003 to 2007 by direct linkage with the Israel National Cancer Registry. Data on diagnostic imaging examinations (CT of chest, abdomen, or pelvis, unspecified sites; X-ray of chest) were extracted during a 5-year follow-up for 226 incident patients. The actual number of CT and X-ray examinations was compared to the National Comprehensive Cancer Network (NCCN) guideline. We tabulated the median with 10th and 90th percentiles (P10, P90) for the number of CTs and X-rays considering histology, stage, and adjuvant strategy. RESULTS The number of abdomen or pelvis CTs for TC patients receiving chemo- or radiotherapy was in accordance with the NCCN guideline. The median of abdomen or pelvis CTs for surveillance patients was 8.5 (P10, P90: 3; 13) for nonseminoma and 5.0 (P10, P90: 5; 13) for seminoma patients compared to 14 to 17 CTs recommended. The number of chest X-rays was lower than recommended in the guideline for all adjuvant strategies. CONCLUSIONS The NCCN guidelines regarding CTs were met for TC patients treated with chemo- or radiotherapy but fell below recommendations for surveillance. Guidelines from 2011 and 2012 were updated in favor of fewer CTs during surveillance. KEY POINTS • The number of CTs followed the NCCN guidelines in patients treated with chemo- or radiotherapy. • Surveillance patients received fewer CTs and X-rays than recommended in the NCCN guidelines from 2005. • The number of applied CT examinations corresponded to a radiation dose that did not substantially raise the lifetime risk for cancer.
Collapse
Affiliation(s)
- Anna-Therese Lehnich
- Center of Clinical Epidemiology, Institute of Medical Informatics, Biometry and Epidemiology, University Hospital of Essen, Hufelandstr. 55, 45147, Essen, Germany.
| | - Carsten Rusner
- Radiology Office, Hospital St. Elisabeth and St. Barbara, Mauerstraße 5, 06110, Halle, Germany
| | - Gabriel Chodick
- Sackler Faculty of Medicine, Tel Aviv University, P.O.B 39040, Ramat Aviv, 69978, Tel Aviv, Israel
- Maccabitech, Maccabi Healthcare Services, Ottoman Society No. 227/99, Of 27 Hamered Street, Tel Aviv, Israel
| | - Rachel Katz
- Maccabitech, Maccabi Healthcare Services, Ottoman Society No. 227/99, Of 27 Hamered Street, Tel Aviv, Israel
| | - Tal Sella
- Maccabitech, Maccabi Healthcare Services, Ottoman Society No. 227/99, Of 27 Hamered Street, Tel Aviv, Israel
- Department of Medical Oncology, Dana-Faber Cancer Institute, 450 Brookline Ave, Boston, MA, USA
| | - Andreas Stang
- Center of Clinical Epidemiology, Institute of Medical Informatics, Biometry and Epidemiology, University Hospital of Essen, Hufelandstr. 55, 45147, Essen, Germany
- School of Public Health, Department of Epidemiology, Boston University, 715 Albany Street, Talbot Building, Boston, MA, 02118, USA
- German Consortium for Translational Cancer Research (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, University Hospital of Essen, Hufelandstr. 55, 45147, Essen, Germany
| |
Collapse
|
30
|
Beyer J. Prognostic factors in metastatic germ-cell cancer. Andrology 2019; 7:475-478. [PMID: 30969027 DOI: 10.1111/andr.12615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/27/2019] [Accepted: 03/04/2019] [Indexed: 01/06/2023]
Abstract
Determining stage and anticipating prognosis are the two crucial steps after the diagnosis of a germ-cell cancer that both guide subsequent treatment decisions. This review focuses on metastatic disease, which means germ-cell cancers beyond stage I limited to the testis. In the past, major centers developed separate staging and prognostic classifications for metastatic germ-cell cancer, which heavily impaired communication and comparisons across clinical trials. More recently, the classification system of the International Germ-Cell Cancer Collaborative Group (IGCCCG) has found acceptance worldwide and is currently being updated.
Collapse
Affiliation(s)
- J Beyer
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
31
|
Lanciotti L, Cofini M, Leonardi A, Bertozzi M, Penta L, Esposito S. Different Clinical Presentations and Management in Complete Androgen Insensitivity Syndrome (CAIS). INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16071268. [PMID: 30970592 PMCID: PMC6480640 DOI: 10.3390/ijerph16071268] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/01/2019] [Accepted: 04/04/2019] [Indexed: 01/08/2023]
Abstract
Complete androgen insensitivity syndrome (CAIS) is an X-linked recessive genetic disorder resulting from maternally inherited or de novo mutations involving the androgen receptor gene, situated in the Xq11-q12 region. The diagnosis is based on the presence of female external genitalia in a 46, XY human individual, with normally developed but undescended testes and complete unresponsiveness of target tissues to androgens. Subsequently, pelvic ultrasound or magnetic resonance imaging (MRI) could be helpful in confirming the absence of Mullerian structures, revealing the presence of a blind-ending vagina and identifying testes. CAIS management still represents a unique challenge throughout childhood and adolescence, particularly regarding timing of gonadectomy, type of hormonal therapy, and psychological concerns. Indeed this condition is associated with an increased risk of testicular germ cell tumour (TGCT), although TGCT results less frequently than in other disorders of sex development (DSD). Furthermore, the majority of detected tumoral lesions are non-invasive and with a low probability of progression into aggressive forms. Therefore, histological, epidemiological, and prognostic features of testicular cancer in CAIS allow postponing of the gonadectomy until after pubertal age in order to guarantee the initial spontaneous pubertal development and avoid the necessity of hormonal replacement therapy (HRT) induction. However, HRT is necessary after gonadectomy in order to prevent symptoms of hypoestrogenism and to maintain secondary sexual features. This article presents differential clinical presentations and management in patients with CAIS to emphasize the continued importance of standardizing the clinical and surgical approach to this disorder.
Collapse
Affiliation(s)
- Lucia Lanciotti
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, 06132 Perugia, Italy.
| | - Marta Cofini
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, 06132 Perugia, Italy.
| | - Alberto Leonardi
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, 06132 Perugia, Italy.
| | - Mirko Bertozzi
- Pediatric Surgery, Azienda Ospedaliera Santa Maria della Misericordia, 20122 Perugia, Italy.
| | - Laura Penta
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, 06132 Perugia, Italy.
| | - Susanna Esposito
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, 06132 Perugia, Italy.
| |
Collapse
|
32
|
Karim S, Wei X, Leveridge MJ, Siemens DR, Robinson AG, Bedard PL, Booth CM. Delivery of chemotherapy for testicular cancer in routine practice: A population-based study. Urol Oncol 2019; 37:183.e17-183.e24. [DOI: 10.1016/j.urolonc.2018.10.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/15/2018] [Accepted: 10/26/2018] [Indexed: 11/27/2022]
|
33
|
Sponholz S, Trainer S, Schirren M, Schirren J. Resection of retrocrural germ cell tumor metastases: Two surgical approaches. J Thorac Cardiovasc Surg 2019; 157:2482-2489. [PMID: 30879726 DOI: 10.1016/j.jtcvs.2019.01.119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 01/17/2019] [Accepted: 01/22/2019] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The goals of retrocrural metastasectomy are complete resection with preservation of the diaphragmatic function while avoiding phrenic nerve injury and spinal cord ischemia. We describe 2 approaches for metastasectomy depending on the pattern of metastases. METHODS Between 1999 and 2017, 44 patients underwent 50 retrocrural metastasectomies. In case of lower retrocrural, bilateral retrocrural, and or additional retroperitoneal and abdominal metastases, an abdominal approach with mobilization of the liver and the kidney followed by longitudinal incision of the diaphragmatic crus was performed. In case of upper retrocrural metastases and additional thoracic disease, a thoracic approach was performed. The Kaplan-Meier method and log-rank test were used to analyze survival and prognosticators. RESULTS The minor morbidity, major morbidity, and mortality were 16.6%, 0%, and 0% for the abdominal approach, respectively, and 15.4%, 3.8%, and 0% for the thoracic approach. There was no phrenic nerve palsy, diaphragmatic hernia, or spinal cord ischemia. Additional retroperitoneal, mediastinal, pulmonary, or further resection was necessary in 10, 25, 9, and 6 cases, respectively. In all cases, a R0 resection was achieved. The 15-year survival rate was 95%. CONCLUSIONS Depending on the pattern of metastases, a complete retrocrural metastasectomy with low morbidity and without mortality by thoracic or abdominal approach is possible. Both approaches preserve diaphragmatic function. Furthermore, the lateral abdominal approach provides a good view and might lead to less tension at the spinal arteries and therefore might reduce the risk of paresis. Good long-term survival is achievable. These patients should be operated on in specialized centers.
Collapse
Affiliation(s)
- Stefan Sponholz
- Department of Thoracic Surgery, Agaplesion Markus Krankenhaus, Frankfurt, Germany.
| | - Stephan Trainer
- Department of Thoracic Surgery, Agaplesion Markus Krankenhaus, Frankfurt, Germany
| | - Moritz Schirren
- Department of Thoracic Surgery, Agaplesion Markus Krankenhaus, Frankfurt, Germany
| | - Joachim Schirren
- Department of Thoracic Surgery, Agaplesion Markus Krankenhaus, Frankfurt, Germany
| |
Collapse
|
34
|
Terbuch A, Adiprasito JB, Stiegelbauer V, Seles M, Klec C, Pichler GP, Resel M, Posch F, Lembeck AL, Stöger H, Szkandera J, Pummer K, Bauernhofer T, Hutterer GC, Gerger A, Stotz M, Pichler M. MiR-371a-3p Serum Levels Are Increased in Recurrence of Testicular Germ Cell Tumor Patients. Int J Mol Sci 2018; 19:ijms19103130. [PMID: 30321995 PMCID: PMC6213366 DOI: 10.3390/ijms19103130] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 12/22/2022] Open
Abstract
Metastatic testicular germ cell tumors (TGCTs) are a potentially curable disease by administration of risk-adapted cytotoxic chemotherapy. Nevertheless, a disease-relapse after curative chemotherapy needs more intensive salvage chemotherapy and significantly worsens the prognosis of TGCT patients. Circulating tumor markers (β-subunit of human chorionic gonadotropin (β-HCG), alpha-Fetoprotein (AFP), and Lactate Dehydrogenase (LDH)) are frequently used for monitoring disease recurrence in TGCT patients, though they lack diagnostic sensitivity and specificity. Increasing evidence suggests that serum levels of stem cell-associated microRNAs (miR-371a-3p and miR-302/367 cluster) are outperforming the traditional tumor markers in terms of sensitivity to detect newly diagnosed TGCT patients. The aim of this study was to investigate whether these miRNAs are also informative in detection of disease recurrence in TGCT patients after curative first line therapy. For this purpose, we measured the serum levels of miR-371a-3p and miR-367 in 52 samples of ten TGCT patients at different time points during disease relapse and during salvage chemotherapy. In our study, miR-371a-3p levels in serum samples with proven disease recurrence were 13.65 fold higher than levels from the same patients without evidence of disease (p = 0.014). In contrast, miR-367 levels were not different in these patient groups (p = 0.985). In conclusion, miR-371a-3p is a sensitive and potentially novel biomarker for detecting disease relapse in TGCT patients. This promising biomarker should be investigated in further large prospective trials.
Collapse
Affiliation(s)
- Angelika Terbuch
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria.
- Drug Development Unit, Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK.
| | - Jan B Adiprasito
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria.
- Research Unit of Non-Coding RNA and Genome Editing in Cancer, Medical University of Graz, 8036 Graz, Austria.
| | - Verena Stiegelbauer
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria.
- Research Unit of Non-Coding RNA and Genome Editing in Cancer, Medical University of Graz, 8036 Graz, Austria.
| | - Maximilian Seles
- Department of Urology, Medical University of Graz, 8036 Graz, Austria.
| | - Christiane Klec
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria.
- Research Unit of Non-Coding RNA and Genome Editing in Cancer, Medical University of Graz, 8036 Graz, Austria.
| | - Georg P Pichler
- Department of Urology, Medical University of Graz, 8036 Graz, Austria.
| | - Margit Resel
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria.
| | - Florian Posch
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria.
- Research Unit of Non-Coding RNA and Genome Editing in Cancer, Medical University of Graz, 8036 Graz, Austria.
| | - Anna L Lembeck
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria.
| | - Herbert Stöger
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria.
| | - Joanna Szkandera
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria.
| | - Karl Pummer
- Department of Urology, Medical University of Graz, 8036 Graz, Austria.
| | - Thomas Bauernhofer
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria.
- Center for Biomarker Research in Medicine (CBmed), 8036 Graz, Austria.
| | - Georg C Hutterer
- Department of Urology, Medical University of Graz, 8036 Graz, Austria.
| | - Armin Gerger
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria.
- Center for Biomarker Research in Medicine (CBmed), 8036 Graz, Austria.
| | - Michael Stotz
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria.
| | - Martin Pichler
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria.
- Research Unit of Non-Coding RNA and Genome Editing in Cancer, Medical University of Graz, 8036 Graz, Austria.
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
35
|
Cools M, Wolffenbuttel KP, Hersmus R, Mendonca BB, Kaprová J, Drop SLS, Stoop H, Gillis AJM, Oosterhuis JW, Costa EMF, Domenice S, Nishi MY, Wunsch L, Quigley CA, T'Sjoen G, Looijenga LHJ. Malignant testicular germ cell tumors in postpubertal individuals with androgen insensitivity: prevalence, pathology and relevance of single nucleotide polymorphism-based susceptibility profiling. Hum Reprod 2018; 32:2561-2573. [PMID: 29121256 DOI: 10.1093/humrep/dex300] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 09/14/2017] [Indexed: 01/29/2023] Open
Abstract
STUDY QUESTION What is the prevalence of malignant testicular germ cell tumors (TGCT) and its precursors, (pre-) germ cell neoplasia in situ (GCNIS), in late teenagers and adults who have androgen insensitivity syndrome (AIS) and the impact of an individual's genetic susceptibility to development of TGCT? SUMMARY ANSWER No GCNIS or TGCT was diagnosed, but pre-GCNIS was identified in 14 and 10% of complete and partial AIS patients, respectively, and was associated with a higher genetic susceptibility score (GSS), with special attention for KITLG (rs995030) and ATFZIP (rs2900333). WHAT IS KNOWN ALREADY Many adult women with AIS decline prophylactic gonadectomy, while data regarding the incidence, pathophysiology and outcomes of TGCT in postpubertal individuals with AIS are lacking. The relevance of genetic factors, such as single nucleotide polymorphisms (SNPs), in predisposing AIS individuals to TGCT is unknown. STUDY DESIGN, SIZE, DURATION This multicenter collaborative study on prophylactically removed gonadal tissue was conducted in a pathology lab specialized in germ cell tumor biology. PARTICIPANTS/MATERIALS, SETTING, METHODS Material from 52 postpubertal individuals with molecularly confirmed AIS (97 gonadal samples) was included; the median age at surgery was 17.5 (14-54) years. Immunohistochemical studies and high-throughput profiling of 14 TGCT-associated SNPs were performed. The main outcome measures were the prevalence of pre-GCNIS, GCNIS and TGCT, and its correlation with a GSS, developed based on the results of recent genome-wide association studies. MAIN RESULTS AND ROLE OF CHANCE The earliest recognizable change preceding GCNIS, referred to as pre-GCNIS, was present in 14% of individuals with complete and 10% of those with partial AIS at a median age of 16 years. No GCNIS or invasive TGCT were found. The median GSS was significantly greater for those with, compared to those without, pre-GCNIS (P = 0.01), with an overlap between groups. Our data suggest important roles for risk alleles G at KITLG (rs995030) and C at ATFZIP (rs2900333), among the 14 studied TGCT-associated SNPs. LARGE SCALE DATA N/A. LIMITATIONS REASONS FOR CAUTION A limited number of cases were included. WIDER IMPLICATIONS OF THE FINDINGS Our data suggest that the prevalence of pre-GCNIS in individuals with AIS beyond puberty is around 15%. Genetic susceptibility likely contributes to pre-GCNIS development in AIS but factors related to malignant progression remain unclear. Although data in older patients remain scarce, malignant progression appears to be a rare event, although the natural history of the premalignant lesion remains unknown. Therefore, the practice of routine prophylactic gonadectomy in adults with AIS appears questionable and the patient's preference, after having been fully informed, should be decisive in this matter. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by research grants from the Research Foundation Flanders (FWO) (to M.C.), the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq G0D6713N) (to B.B.M. and M.C.) and the European Society for Pediatric Endocrinology (ESPE), granted by Novo Nordisk AB (to J.K.). There are no competing interests.
Collapse
Affiliation(s)
- M Cools
- Pediatrics and Genetics, Ghent University and Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
| | - K P Wolffenbuttel
- Department of Pediatric Urology, Sophia Children's Hospital and Erasmus Medical Center, Rotterdam, The Netherlands
| | - R Hersmus
- Laboratory for Experimental Patho-Oncology, Josephine Nefkens Institute and Erasmus Medical Center, Rotterdam, The Netherlands
| | - B B Mendonca
- Department of Endocrinology, Hormone and Molecular Genetics Laboratory, LIM/42 Clinicas Hospital; University of Sao Paulo, Sao Paulo, Brazil
| | - J Kaprová
- Laboratory for Experimental Patho-Oncology, Josephine Nefkens Institute and Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Pediatrics, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - S L S Drop
- Department of Pediatric Endocrinology, Sophia Children's Hospital and Erasmus Medical Center, Rotterdam, The Netherlands
| | - H Stoop
- Laboratory for Experimental Patho-Oncology, Josephine Nefkens Institute and Erasmus Medical Center, Rotterdam, The Netherlands
| | - A J M Gillis
- Laboratory for Experimental Patho-Oncology, Josephine Nefkens Institute and Erasmus Medical Center, Rotterdam, The Netherlands
| | - J W Oosterhuis
- Laboratory for Experimental Patho-Oncology, Josephine Nefkens Institute and Erasmus Medical Center, Rotterdam, The Netherlands
| | - E M F Costa
- Department of Endocrinology, Hormone and Molecular Genetics Laboratory, LIM/42 Clinicas Hospital; University of Sao Paulo, Sao Paulo, Brazil
| | - S Domenice
- Department of Endocrinology, Hormone and Molecular Genetics Laboratory, LIM/42 Clinicas Hospital; University of Sao Paulo, Sao Paulo, Brazil
| | - M Y Nishi
- Department of Endocrinology, Hormone and Molecular Genetics Laboratory, LIM/42 Clinicas Hospital; University of Sao Paulo, Sao Paulo, Brazil
| | - L Wunsch
- Department of Pediatric Urology, Universitätsklinikum Schleswig-Holstein and Universität zu Lübeck, Lübeck, Germany
| | - C A Quigley
- Pediatric Endocrinology, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - G T'Sjoen
- Internal Medicine, Ghent University and Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
| | - L H J Looijenga
- Laboratory for Experimental Patho-Oncology, Josephine Nefkens Institute and Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
36
|
Risk of diabetes after para-aortic radiation for testicular cancer. Br J Cancer 2018; 119:901-907. [PMID: 30297773 PMCID: PMC6189211 DOI: 10.1038/s41416-018-0248-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 07/19/2018] [Accepted: 08/02/2018] [Indexed: 01/04/2023] Open
Abstract
Background While the risk of diabetes is increased following radiation exposure to the pancreas among childhood cancer survivors, its association among testicular cancer (TC) survivors has not been investigated. Methods Diabetes risk was studied in 2998 1-year TC survivors treated before 50 years of age with orchidectomy with/without radiotherapy between 1976 and 2007. Diabetes incidence was compared with general population rates. Treatment-specific risk of diabetes was assessed using a case–cohort design. Results With a median follow-up of 13.4 years, 161 TC survivors were diagnosed with diabetes. Diabetes risk was not increased compared to general population rates (standardised incidence ratios (SIR): 0.9; 95% confidence interval (95% CI): 0.7–1.1). Adjusted for age, para-aortic radiotherapy was associated with a 1.66-fold (95% CI: 1.05–2.62) increased diabetes risk compared to no radiotherapy. The excess hazard increased with 0.31 with every 10 Gy increase in the prescribed radiation dose (95% CI: 0.11–0.51, P = 0.003, adjusted for age and BMI); restricted to irradiated patients the excess hazard increased with 0.33 (95% CI: −0.14 to 0.81, P = 0.169) with every 10 Gy increase in radiation dose. Conclusion Compared to surgery only, para-aortic irradiation is associated with increased diabetes risk among TC survivors.
Collapse
|
37
|
Groot HJ, Lubberts S, de Wit R, Witjes JA, Kerst JM, de Jong IJ, Groenewegen G, van den Eertwegh AJ, Poortmans PM, Klümpen HJ, van den Berg HA, Smilde TJ, Vanneste BG, Aarts MJ, Incrocci L, van den Bergh AC, Jóźwiak K, van den Belt-Dusebout AW, Horenblas S, Gietema JA, van Leeuwen FE, Schaapveld M. Risk of Solid Cancer After Treatment of Testicular Germ Cell Cancer in the Platinum Era. J Clin Oncol 2018; 36:2504-2513. [DOI: 10.1200/jco.2017.77.4174] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Purpose Testicular cancer (TC) treatment increases risk of subsequent malignant neoplasms (SMNs). It is unknown whether changes in TC treatment over time have affected SMN risk. Methods Solid SMN risk was evaluated in a multicenter cohort comprising 5,848 1-year survivors treated for TC before age 50 years between 1976 and 2007. SMN incidence was compared with cancer incidence in the general population. Treatment-specific risks were assessed using multivariable regression in a case-cohort design. Results After a median follow-up of 14.1 years, 350 solid SMNs were observed, translating into a 1.8-fold (95% CI, 1.6-2.0) increased risk compared with general population rates. Solid SMN risk was increased in patients with seminoma and those with nonseminoma (standardized incidence ratio, 1.52 and 2.21, respectively). Patients with nonseminoma experienced increased risk of SMNs of the thyroid, lung, stomach, pancreas, colon, and bladder and of melanoma and soft tissue sarcoma, whereas those with seminoma experienced increased risk of SMNs of the small intestine, pancreas, and urinary bladder. The 25-year cumulative incidence of solid SMNs was 10.3% (95% CI, 9.0% to 11.6%). In multivariable analysis, platinum-based chemotherapy was associated with increased risk of a solid SMN (hazard ratio [HR], 2.40; 95% CI, 1.58 to 3.62), colorectal SMN (HR, 3.85; 95% CI, 1.67 to 8.92), and noncolorectal GI SMN (HR, 5.00; 95% CI, 2.28 to 10.95). Receipt of platinum 400 to 499 and ≥ 500 mg/m2 increased solid SMN risk compared with surgery only (HR, 2.43; 95% CI, 1.40 to 4.23 and HR, 2.42; 95% CI, 1.50 to 3.90, respectively), whereas risk was not significantly increased with lower doses (HR, 1.75; 95% CI, 0.90 to 3.43). The HR of a GI SMN increased by 53% (95% CI, 26% to 80%) per 100 mg/m2 of platinum-containing chemotherapy. The HR of an infradiaphragmatic SMN increased by 8% per Gray of radiation dose administered (95% CI, 6% to 9%; P < .001). Conclusion Radiotherapy and platinum-containing chemotherapy are associated with increased solid SMN risk, specifically with GI SMNs.
Collapse
Affiliation(s)
- Harmke J. Groot
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Sjoukje Lubberts
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Ronald de Wit
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Johannes A. Witjes
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Jan Martijn Kerst
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Igle J. de Jong
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Gerard Groenewegen
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Alfons J.M. van den Eertwegh
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Philip M. Poortmans
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Heinz-Josef Klümpen
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Hetty A. van den Berg
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Tineke J. Smilde
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Ben G.L. Vanneste
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Maureen J. Aarts
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Luca Incrocci
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Alfons C.M. van den Bergh
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Katarzyna Jóźwiak
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Alexandra W. van den Belt-Dusebout
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Simon Horenblas
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Jourik A. Gietema
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Flora E. van Leeuwen
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| | - Michael Schaapveld
- Harmke J. Groot, Jan Martijn Kerst, Katarzyna Jóźwiak, Alexandra W. van den Belt-Dusebout, Simon Horenblas, Flora E. van Leeuwen, and Michael Schaapveld, Netherlands Cancer Institute; Alfons J.M. van den Eertwegh, Vrije Universiteit Medical Center; Heinz-Josef Klümpen, Academic Medical Center, Amsterdam; Sjoukje Lubberts, Igle J. de Jong, Alfons C.M. van den Bergh, and Jourik A. Gietema, University Medical Center Groningen, University of Groningen, Groningen; Ronald de Wit and Luca Incrocci, Erasmus
| |
Collapse
|
38
|
Mansouri N, Boujelbène N, Abbes I, Mezghani N, Doghri R, Charfi L, Driss M, Chkili R, Mrad K. A rare case of a brain metastasis in a 4year old child. Ann Pathol 2018; 39:14-17. [PMID: 30078515 DOI: 10.1016/j.annpat.2018.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 05/20/2018] [Accepted: 06/16/2018] [Indexed: 10/28/2022]
Affiliation(s)
- Nada Mansouri
- Pathology Department, Military Hospital, Tunis, Tunisia.
| | | | - Imen Abbes
- Pathology Department, Salah Azaiez Institute, Tunis, Tunisia.
| | | | - Raoudha Doghri
- Pathology Department, Salah Azaiez Institute, Tunis, Tunisia.
| | - Lamia Charfi
- Pathology Department, Salah Azaiez Institute, Tunis, Tunisia.
| | - Maha Driss
- Pathology Department, Salah Azaiez Institute, Tunis, Tunisia.
| | - Ridha Chkili
- Neurosugery Department, Military Hospital, Tunis, Tunisia.
| | - Karima Mrad
- Pathology Department, Salah Azaiez Institute, Tunis, Tunisia.
| |
Collapse
|
39
|
Leveridge MJ, Siemens DR, Brennan K, Izard JP, Karim S, An H, Mackillop WJ, Booth CM. Temporal trends in management and outcomes of testicular cancer: A population-based study. Cancer 2018; 124:2724-2732. [PMID: 29660851 DOI: 10.1002/cncr.31390] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 01/31/2018] [Accepted: 02/20/2018] [Indexed: 11/09/2022]
Abstract
BACKGROUND Treatment guidelines for early-stage testicular cancer have increasingly recommended de-escalation of therapy with surveillance strategies. This study was designed to describe temporal trends in routine clinical practice and to determine whether de-escalation of therapy is associated with inferior survival in the general population. METHODS The Ontario Cancer Registry was linked to electronic records of treatment to identify all patients diagnosed with testicular cancer treated with orchiectomy in Ontario during 2000-2010. Treatment after orchiectomy was classified as radiotherapy (RT), retroperitoneal lymph node dissection (RPLND), chemotherapy, or none. Surveillance was defined as no identified treatment within 90 days of orchiectomy. Overall survival (OS) and cancer-specific survival (CSS) were measured from the date of orchiectomy. RESULTS The study population included 1564 and 1086 cases of seminomas and nonseminoma germ cell tumors (NSGCTs), respectively. Among patients with seminomas, there was a significant increase in the proportion of patients with no treatment within 90 days of orchiectomy (from 56% to 84%; P < .001); the use of RT decreased over time (from 38% to 8%; P < .001); and the use of chemotherapy remained stable (from 6% to 9%; P = .289). Practice patterns 90 days after orchiectomy remained stable over time among patients with NSGCTs: from 51% to 57% for no treatment (P = .435), from 43% to 43% for chemotherapy (P = .336), and from 9% to 3% for RPLND (P = .476). The OS rates for the entire cohort at 5 and 10 years were 97% and 96%, respectively; the CSS rates were 98% and 98%, respectively. There was no significant change in OS or CSS for patients with seminomas or NSGCTs during the study period. CONCLUSIONS There has been substantial de-escalation in the treatment of testicular cancer in routine practice since 2000. Long-term survival in routine practice is excellent and has not decreased with the uptake of surveillance strategies. Cancer 2018;124:2724-2732. © 2018 American Cancer Society.
Collapse
Affiliation(s)
- Michael J Leveridge
- Department of Urology, Queen's University, Kingston, Ontario, Canada.,Department of Oncology, Queen's University, Kingston, Ontario, Canada
| | - D Robert Siemens
- Department of Urology, Queen's University, Kingston, Ontario, Canada.,Department of Oncology, Queen's University, Kingston, Ontario, Canada
| | - Kelly Brennan
- Division of Cancer Care and Epidemiology, Queen's University Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
| | - Jason P Izard
- Department of Urology, Queen's University, Kingston, Ontario, Canada.,Department of Oncology, Queen's University, Kingston, Ontario, Canada
| | - Safiya Karim
- Department of Oncology, Queen's University, Kingston, Ontario, Canada.,Division of Cancer Care and Epidemiology, Queen's University Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
| | - Howard An
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - William J Mackillop
- Department of Oncology, Queen's University, Kingston, Ontario, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,Department of Public Health Sciences, Queen's University, Kingston, Ontario, Canada
| | - Christopher M Booth
- Department of Oncology, Queen's University, Kingston, Ontario, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,Department of Public Health Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
40
|
In Vivo 6-([ 18F]Fluoroacetamido)-1-hexanoicanilide PET Imaging of Altered Histone Deacetylase Activity in Chemotherapy-Induced Neurotoxicity. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:3612027. [PMID: 29755299 PMCID: PMC5884410 DOI: 10.1155/2018/3612027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 01/19/2018] [Accepted: 01/31/2018] [Indexed: 12/26/2022]
Abstract
Background Histone deacetylases (HDACs) regulate gene expression by changing histone deacetylation status. Neurotoxicity is one of the major side effects of cisplatin, which reacts with deoxyribonucleic acid (DNA) and has excellent antitumor effects. Suberoylanilide hydroxamic acid (SAHA) is an HDAC inhibitor with neuroprotective effects against cisplatin-induced neurotoxicity. Purpose We investigated how cisplatin with and without SAHA pretreatment affects HDAC expression/activity in the brain by using 6-([18F]fluoroacetamido)-1-hexanoicanilide ([18F]FAHA) as a positron emission tomography (PET) imaging agent for HDAC IIa. Materials and Methods [18F]FAHA and [18F]fluoro-2-deoxy-2-D-glucose ([18F]FDG) PET studies were done in 24 mice on 2 consecutive days and again 1 week later. The mice were divided into three groups according to drug administration between the first and second imaging sessions (Group A: cisplatin 2 mg/kg, twice; Group B: cisplatin 4 mg/kg, twice; Group C: cisplatin 4 mg/kg, twice, and SAHA 300 mg/kg pretreatment, 4 times). Results The Ki value of [18F]FAHA was increased and the percentage of injected dose/tissue g (% ID/g) of [18F]FDG was decreased in the brains of animals in Groups A and B. The Ki value of [18F]FAHA and % ID/g of [18F]FDG were not significantly different in Group C. Conclusions [18F]FAHA PET clearly showed increased HDAC activity suggestive of cisplatin neurotoxicity in vivo, which was blocked by SAHA pretreatment.
Collapse
|
41
|
Dowling CM, Assel M, Musser JE, Meeks JJ, Sjoberg DD, Bosl G, Motzer R, Bajorin D, Feldman D, Carver BS, Sheinfeld J. Clinical Outcome of Retroperitoneal Lymph Node Dissection after Chemotherapy in Patients with Pure Embryonal Carcinoma in the Orchiectomy Specimen. Urology 2018; 114:133-138. [PMID: 29410311 DOI: 10.1016/j.urology.2018.01.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To determine the pathologic findings and clinical outcome of patients with pure embryonal carcinoma (EC) of the testis who were diagnosed with testis cancer from January 1989 to January 2013 who underwent an orchiectomy, cisplatin-based chemotherapy and a postchemotherapy retroperitoneal lymph node dissection (PC-RPLND). METHODS We compared those patients with 100% EC with those with mixed nonseminomatous germ cell tumor pathology who underwent a PC-RPLND. RESULTS Of 1105 patients who underwent a PC-RPLND, 145 had pure EC. Twenty-six percent of patients presented with metastatic disease outside the retroperitoneum. Patients with mixed histologies tended to have worse International Germ Cell Cancer Collaborative Group risk compared to those with EC at orchiectomy (P = .037). Histology at PC-RPLND revealed fibrosis or necrosis in 76%, mature teratoma in 19% and viable cancer in 4%. Over one-third of the patients had a residual mass of <1 cm prior to RPLND; of whom 15% harbored mature teratoma in PC-RPLND histology. The Kaplan-Meier estimated probability of recurrence at 5 years of follow-up was 3.1% (95% CI 1.2%, 8.0%) for EC histology, 7.3% lower than mixed histology. For cancer-specific mortality, the Kaplan-Meier estimated probability at 5 years was 4.6% (95% CI 3.3%, 6.3%) and 1.7% (95% CI 0.4%, 6.8%) for mixed and pure EC histologies, respectively. CONCLUSION Approximately 20% of patients with pure EC had teratoma at PC-RPLND. We have shown that those with a maximum node size of <1 cm should not be precluded from RPLND.
Collapse
Affiliation(s)
- Catherine M Dowling
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, NY
| | - Melissa Assel
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY
| | - John E Musser
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, NY
| | - Joshua J Meeks
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, NY
| | - Daniel D Sjoberg
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY
| | - George Bosl
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, NY
| | - Robert Motzer
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, NY
| | - Dean Bajorin
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, NY
| | - Darren Feldman
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, NY
| | - Brett S Carver
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, NY
| | - Joel Sheinfeld
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, NY.
| |
Collapse
|
42
|
Terbuch A, Posch F, Partl R, Zurl B, Bauernhofer T, Pichler M, Szkandera J, Hutterer GC, Pummer K, Kapp KS, Stöger H, Gerger A, Stotz M. Risk stratification for febrile neutropenia in patients with testicular germ cell tumors. Cancer Med 2018; 7:508-514. [PMID: 29349917 PMCID: PMC5806095 DOI: 10.1002/cam4.1317] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 12/05/2017] [Accepted: 12/06/2017] [Indexed: 02/05/2023] Open
Abstract
The aim of this study was to detect risk factors for febrile neutropenia (FN) in patients with testicular germ cell tumors (TGCT). In this retrospective cohort study at the Medical University of Graz, we included 413 consecutive TGCT patients who received adjuvant or curative treatment with cisplatin-based chemotherapy. FN occurred in 70 (16.9%) of 413 patients. In univariable logistic regression, higher age (odds ratio (OR) per 5 years = 1.17, 95% CI: 1.02-1.35, P = 0.022), reduced performance status (PS) (OR = 2.73, 1.47-5.06, P = 0.001), seminomatous histology (OR = 2.19, 1.26-3.78, P = 0.005), poor IGCCCG risk class (OR = 4.20, 1.71-10.33, P = 0.002), and prior radiotherapy (pRTX) (OR = 8.98, 2.09-38.61, P = 0.003) were associated with a higher risk of FN. In multivariable analysis adjusting for age and risk classification, only poor PS (OR = 2.06, 1.05-4.03, P = 0.035), seminomatous histology (OR = 2.08, 1.01-4.26, P = 0.047), and pRTX (OR = 7.31, 1.61-33.17, P = 0.010) prevailed. In the subgroup of seminoma patients (n = 104), only pRTX predicted for FN risk (OR = 5.60, 1.24-25.34, P = 0.025). Five of eight seminoma patients with pRTX developed FN (63%), as compared to 22 FN cases (23%) in the 96 seminoma patients without pRTX (P = 0.027). The eight seminoma patients who received pRTX had significantly lower pre-chemo white blood counts (4.7 vs. 6.5 G/L), neutrophil counts (3.2 vs. 4.3 G/L), and platelet counts (185 vs. 272 G/L) than patients without pRTX (all P < 0.0001). TGCT patients with a reduced performance status or who had been previously treated with radiotherapy have an increased risk for neutropenic fever during chemotherapy.
Collapse
Affiliation(s)
- Angelika Terbuch
- Division of OncologyDepartment of Internal MedicineComprehensive Cancer Center GrazMedical University of GrazGrazAustria
- Research Unit Genetic Epidemiology and PharmacogeneticsMedical University of GrazGrazAustria
| | - Florian Posch
- Division of OncologyDepartment of Internal MedicineComprehensive Cancer Center GrazMedical University of GrazGrazAustria
- Research Unit Genetic Epidemiology and PharmacogeneticsMedical University of GrazGrazAustria
| | - Richard Partl
- Department of Therapeutic Radiology and OncologyComprehensive Cancer Center GrazMedical University of GrazGrazAustria
| | - Brigitte Zurl
- Department of Therapeutic Radiology and OncologyComprehensive Cancer Center GrazMedical University of GrazGrazAustria
| | - Thomas Bauernhofer
- Division of OncologyDepartment of Internal MedicineComprehensive Cancer Center GrazMedical University of GrazGrazAustria
- Center for Biomarker Research in Medicine (CBmed)GrazAustria
| | - Martin Pichler
- Division of OncologyDepartment of Internal MedicineComprehensive Cancer Center GrazMedical University of GrazGrazAustria
- Department of Experimental TherapeuticsThe University of TexasMD Anderson Cancer CenterHoustonTexas
| | - Joanna Szkandera
- Division of OncologyDepartment of Internal MedicineComprehensive Cancer Center GrazMedical University of GrazGrazAustria
| | | | - Karl Pummer
- Department of UrologyMedical University of GrazGrazAustria
| | - Karin S. Kapp
- Department of Therapeutic Radiology and OncologyComprehensive Cancer Center GrazMedical University of GrazGrazAustria
| | - Herbert Stöger
- Division of OncologyDepartment of Internal MedicineComprehensive Cancer Center GrazMedical University of GrazGrazAustria
| | - Armin Gerger
- Division of OncologyDepartment of Internal MedicineComprehensive Cancer Center GrazMedical University of GrazGrazAustria
- Research Unit Genetic Epidemiology and PharmacogeneticsMedical University of GrazGrazAustria
- Center for Biomarker Research in Medicine (CBmed)GrazAustria
| | - Michael Stotz
- Division of OncologyDepartment of Internal MedicineComprehensive Cancer Center GrazMedical University of GrazGrazAustria
- Research Unit Genetic Epidemiology and PharmacogeneticsMedical University of GrazGrazAustria
| |
Collapse
|
43
|
Abstract
Germ cell tumors are rare neoplasms that affect young males. Nearly 99% of patients with localized stage I disease and nearly 80% of patients with metastatic disease can be cured. Even patients who relapse following chemotherapy can achieve a long-term survival in approximately 30–40% of cases. The main objective in early stages and in good prognosis patients has changed in recent years, and it has become of major importance to reduce treatment-related morbidity without compromising the excellent long-term survival rate. In poor prognosis patients, there is a correlation between the experience of the treating institution and the long-term clinical outcome of the patients, particularly when the most sophisticated therapies are needed. So far, of utmost importance is the information from updated practice guidelines for the diagnosis and treatment of germ cell tumors. The Italian Germ cell cancer Group (IGG) has developed the following clinical recommendations, which identify the current standards in diagnosis and treatment of germ cell tumors in adult males.
Collapse
|
44
|
Konrade I, Zavorikina J, Fridvalde A, Rots D, Kalere I, Strumfa I, Dambrova M, Gailite L. Novel Variant of the Androgen Receptor Gene in a Patient With Complete Androgen Insensitivity Syndrome and Polyorchidism. Front Endocrinol (Lausanne) 2018; 9:795. [PMID: 30705665 PMCID: PMC6345100 DOI: 10.3389/fendo.2018.00795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 12/18/2018] [Indexed: 11/13/2022] Open
Abstract
Introduction: Complete androgen insensitivity (CAIS) in 65-95% cases is caused by pathogenic allelic variants (mutations) in the gene encoding androgen receptor (AR gene) and is characterized by female phenotype development with a male karyotype (46, XY). Patients are usually diagnosed during puberty and undergo gonadectomy due to increased testicular germ cell tumor risk. Only a few outcomes have been reported in older individuals with postponed gonadectomy. Case presentation: A 48-year-old CAIS patient presented with polyorchidism (four testes) without gonadal malignancies. Genetic testing identified a novel allelic variant in the AR gene [c.2141T>G (p.Phe805Cys)] causing the clinical symptoms. Conclusion: We have described a unique patient with CAIS and polyorchidism without malignancies in her late 40's bearing a novel likely pathogenic variant in the AR gene.
Collapse
Affiliation(s)
- Ilze Konrade
- Department of Internal Medicine, Riga Stradins University, Riga, Latvia
- Department of Endocrinology, Riga East University Hospital, Riga, Latvia
| | - Julija Zavorikina
- Department of Internal Medicine, Riga Stradins University, Riga, Latvia
| | - Aija Fridvalde
- Department of Internal Medicine, Riga Stradins University, Riga, Latvia
| | - Dmitrijs Rots
- Scientific Laboratory of Molecular Genetics, Riga Stradins University, Riga, Latvia
| | - Ieva Kalere
- Department of Pharmacy, Riga Stradins University, Riga, Latvia
| | - Ilze Strumfa
- Department of Pathology, Riga Stradins University, Riga, Latvia
| | - Maija Dambrova
- Department of Human Physiology and Biochemistry, Riga Stradins University, Riga, Latvia
| | - Linda Gailite
- Scientific Laboratory of Molecular Genetics, Riga Stradins University, Riga, Latvia
- *Correspondence: Linda Gailite
| |
Collapse
|
45
|
Therapeutic targeting of extracellular DNA improves the outcome of intestinal ischemic reperfusion injury in neonatal rats. Sci Rep 2017; 7:15377. [PMID: 29133856 PMCID: PMC5684414 DOI: 10.1038/s41598-017-15807-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 11/02/2017] [Indexed: 12/15/2022] Open
Abstract
Thrombosis and inflammation cooperate in the development of intestinal infarction. Recent studies suggest that extracellular DNA released by damaged cells or neutrophils in form of extracellular traps (NETs) contributes to organ damage in experimental models of ischemia-reperfusion injury. Here we compared the therapeutic effects of targeting fibrin or extracellular DNA in intestinal infarction after midgut volvulus in rats. Following iatrogenic midgut volvulus induction for 3 hours, we treated animals with a combination of tissue plasminogen activator (tPA) and low molecular weight heparin (LMWH) to target fibrin or with DNase1 to degrade extracellular DNA. The therapeutic effects of tPA/LMWH and DNase1 were analyzed after 7 days. We observed that both therapeutic interventions ameliorated tissue injury, apoptosis, and oxidative stress in the intestine. DNase1, but not tPA/LMWH, reduced intestinal neutrophil infiltration and histone-myeloperoxidase-complexes, a surrogate marker of NETs, in circulation. Importantly, tPA/LMWH, but not DNase1, interfered with hemostasis as evidenced by a prolonged tail bleeding time. In conclusion, our data suggest that the therapeutic targeting of fibrin and extracellular DNA improves the outcome of midgut volvulus in rats. DNase1 therapy reduces the inflammatory response including NETs without increasing the risk of bleeding. Thus, targeting of extracellular DNA may provide a safe therapy for patients with intestinal infarction in future.
Collapse
|
46
|
Boettcher M, Meier D, Jiménez-Alcázar M, Eschenburg G, Mietzsch S, Vincent D, Klinke M, Trochimiuk M, Appl B, Tiemann B, Bergholz R, Reinshagen K, Fuchs TA. Degradation of Extracellular DNA by DNase1 Significantly Reduces Testicular Damage After Testicular Torsion in Rats. Urology 2017; 109:223.e1-223.e7. [PMID: 28774773 DOI: 10.1016/j.urology.2017.07.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/24/2017] [Accepted: 07/20/2017] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To examine the effects of DNase1 treatment on testicular damage after testicular torsion (TT). It has been demonstrated that TT induces thrombus formation and that anticoagulation significantly reduces testicular damage after TT. It was hypothesized that these thrombi are dependent on neutrophil extracellular traps (NETs) and thus NETs disintegration would reduce testicular cell damage. METHODS A sham operation was performed in 10 rats. Thirty-four rats underwent induction of iatrogenic TT for 3 hours. After de-torsion and randomization, 24 rats received DNase1 or inactivated DNase1. The following parameters were assessed: testicular damage via Cosentino grading; spermatogenesis via Johnsen score; stem cell factor and c-Kit, apoptosis via Bax, Bcl2, Terminal deoxynucleotidyl transferase (TdT) dUTP Nick-End Labeling assay, and cleaved caspase3 staining; oxidative stress via superoxide dismutase, catalase, glutathione peroxidase, and malondialdehyde; neutrophil recruitment via myeloperoxidase and neutrophil elastase staining; and NET formation via cell-free DNA. RESULTS Forty-three rats were included in the study. Subjects treated with DNase1 showed significantly less cellular damage, oxidative stress, and apoptosis. Further, DNase1-treated rats demonstrated a significant improvement of spermatogenesis, compared with the controls. CONCLUSION The results of the study indicate that thrombus formation during TT is quite likely NET associated, and that dissolution of cell-free DNA (including NETs) significantly improves testicular damage in rats. As treatment with DNase1 reduced apoptosis, oxidative stress, and inflammation, without adversely affecting coagulation, it might be a suitable treatment for (neonatal) TT and ought to be evaluated in humans.
Collapse
Affiliation(s)
- Michael Boettcher
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Dennis Meier
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Miguel Jiménez-Alcázar
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Georg Eschenburg
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Mietzsch
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Deirdre Vincent
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michaela Klinke
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Magdalena Trochimiuk
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Birgit Appl
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bastian Tiemann
- Department of Experimental Animal Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Robert Bergholz
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias A Fuchs
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
47
|
Quezada Bautista A, Lara Bejarano J, García García J, Ortega-García O, Bautista Hernández M. Relapse and gastrointestinal toxicity associated with radiotherapy treatment in stage I seminoma patients. REVISTA MÉDICA DEL HOSPITAL GENERAL DE MÉXICO 2017. [DOI: 10.1016/j.hgmx.2016.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
48
|
Bezan A, Posch F, Ploner F, Bauernhofer T, Pichler M, Szkandera J, Hutterer GC, Pummer K, Gary T, Samonigg H, Beyer J, Winder T, Hermanns T, Fankhauser CD, Gerger A, Stotz M. Risk stratification for venous thromboembolism in patients with testicular germ cell tumors. PLoS One 2017; 12:e0176283. [PMID: 28430804 PMCID: PMC5400272 DOI: 10.1371/journal.pone.0176283] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 04/07/2017] [Indexed: 11/18/2022] Open
Abstract
Background Patients with testicular germ cell tumors (TGCT) have an increased risk for venous thromboembolism (VTE). We identified risk factors for VTE in this patient cohort and developed a clinical risk model. Methods In this retrospective cohort study at the Medical University of Graz we included 657 consecutive TGCT patients across all clinical stages. A predictive model for VTE was developed and externally validated in 349 TGCT patients treated at the University Hospital Zurich. Results Venous thromboembolic events occurred in 34 (5.2%) patients in the Graz cohort. In univariable competing risk analysis, higher clinical stage (cS) and a retroperitoneal lymphadenopathy (RPLN) were the strongest predictors of VTE (p<0.0001). As the presence of a RPLN with more than 5cm in greatest dimension without coexisting visceral metastases is classified as cS IIC, we constructed an empirical VTE risk model with the following four categories (12-month-cumulative incidence): cS IA-B 8/463 patients (1.7%), cS IS-IIB 5/86 patients (5.9%), cS IIC 3/21 patients (14.3%) and cS IIIA-C 15/70 patients (21.4%). This risk model was externally validated in the Zurich cohort (12-month-cumulative incidence): cS IA-B (0.5%), cS IS-IIB (6.0%), cS IIC (11.1%) and cS IIIA-C (19.1%). Our model had a significantly higher discriminatory performance than a previously published classifier (RPLN-VTE-risk-classifier) which is based on the size of RPLN alone (AUC-ROC: 0.75 vs. 0.63, p = 0.007). Conclusions According to our risk stratification, TGCT patients with cS IIC and cS III disease have a very high risk of VTE and may benefit from primary thromboprophylaxis for the duration of chemotherapy.
Collapse
Affiliation(s)
- Angelika Bezan
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Research Unit Genetic Epidemiology and Pharmacogenetics, Medical University of Graz, Graz, Austria
| | - Florian Posch
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Research Unit Genetic Epidemiology and Pharmacogenetics, Medical University of Graz, Graz, Austria
| | - Ferdinand Ploner
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Thomas Bauernhofer
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Martin Pichler
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Department of Experimental Therapeutics, The University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Joanna Szkandera
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | | | - Karl Pummer
- Department of Urology, Medical University of Graz, Graz, Austria
| | - Thomas Gary
- Division of Angiology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | - Hellmut Samonigg
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Joerg Beyer
- Department of Oncology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Thomas Winder
- Department of Oncology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Thomas Hermanns
- Department of Urology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Christian D. Fankhauser
- Department of Urology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
- * E-mail: (CDF); (AG)
| | - Armin Gerger
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Research Unit Genetic Epidemiology and Pharmacogenetics, Medical University of Graz, Graz, Austria
- Center for Biomarker Research in Medicine (CBmed), Graz, Austria
- * E-mail: (CDF); (AG)
| | - Michael Stotz
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Research Unit Genetic Epidemiology and Pharmacogenetics, Medical University of Graz, Graz, Austria
| |
Collapse
|
49
|
Hirakawa H, Nakashima C, Nakamura T, Masuda M, Funakoshi T, Nakagawa S, Horimatsu T, Matsubara K, Muto M, Kimura S, Sueoka-Aragane N. Chemotherapy for primary mediastinal yolk sac tumor in a patient undergoing chronic hemodialysis: a case report. J Med Case Rep 2017; 11:43. [PMID: 28202048 PMCID: PMC5312436 DOI: 10.1186/s13256-017-1213-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 01/17/2017] [Indexed: 11/21/2022] Open
Abstract
Background The safety and efficacy of chemotherapy for patients undergoing concomitant hemodialysis have not been fully established and optimal doses of anti-cancer drugs and best timing of hemodialysis remains unclear. Although chemosensitive cancers, such as germ cell tumors, treated with chemotherapy should have sufficient dose intensity maintained to achieve the desired effect, many patients with cancer undergoing hemodialysis might be under-treated because the pharmacokinetics of anti-cancer drugs in such patients remains unknown. Case presentation We describe a 31-year-old Japanese man with a mediastinal yolk sac tumor treated with surgery followed by five cycles of chemotherapy containing cisplatin and etoposide while concomitantly undergoing hemodialysis. The doses of these agents used in the first cycle were 50% of the standard dose of cisplatin (10 mg/m2) and 60% of the standard dose of etoposide (60 mg/m2) on days 1 through to 5; the doses were subsequently escalated to 75% with both agents. Hemodialysis was started 1 hour after infusions of these agents. Severe hematological toxicities were observed despite successful treatment. During treatment with concurrent hemodialysis, pharmacokinetic analysis of cisplatin was performed and its relationship with adverse effects was assessed. Compared with patients with normal renal function, the maximum drug concentration was higher, and concentration increased in the interval between hemodialysis and the subsequent cisplatin infusion, resulting in a higher area under the curve despite a reduction in the dose to 75% of the standard regimen. Conclusions Because of the altered pharmacokinetics pharmacodynamics status of patients with renal dysfunction undergoing hemodialysis, pharmacokinetics pharmacodynamics analysis is deemed to be helpful for effective and safe management of chemotherapy in patients undergoing hemodialysis.
Collapse
Affiliation(s)
- Haruki Hirakawa
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Naoko Sueoka-Aragane, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Chiho Nakashima
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Naoko Sueoka-Aragane, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Tomomi Nakamura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Naoko Sueoka-Aragane, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Masanori Masuda
- Department of Pathology, Faculty of Medicine, Saga University Hospital, Saga, Japan
| | - Taro Funakoshi
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Takahiro Horimatsu
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuo Matsubara
- Department of Pharmacy, Kyoto University Hospital, Kyoto, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Naoko Sueoka-Aragane, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Naoko Sueoka-Aragane
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Naoko Sueoka-Aragane, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan.
| |
Collapse
|
50
|
Jostes S, Nettersheim D, Fellermeyer M, Schneider S, Hafezi F, Honecker F, Schumacher V, Geyer M, Kristiansen G, Schorle H. The bromodomain inhibitor JQ1 triggers growth arrest and apoptosis in testicular germ cell tumours in vitro and in vivo. J Cell Mol Med 2016; 21:1300-1314. [PMID: 28026145 PMCID: PMC5487916 DOI: 10.1111/jcmm.13059] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/15/2016] [Indexed: 12/21/2022] Open
Abstract
Type II testicular germ cell cancers (TGCT) are the most frequently diagnosed tumours in young men (20–40 years) and are classified as seminoma or non‐seminoma. TGCTs are commonly treated by orchiectomy and chemo‐ or radiotherapy. However, a subset of metastatic non‐seminomas (embryonal carcinomas) displays only incomplete remission or relapse and requires novel treatment options. Recent studies have shown effective application of the small‐molecule inhibitor JQ1 in tumour therapy, which interferes with the function of ‘bromodomain and extraterminal (BET)’ proteins. JQ1‐treated TGCT cell lines display up‐regulation of genes indicative for DNA damage and cellular stress response and induce cell cycle arrest. Embryonal carcinoma (EC) cell lines, which presented as JQ1 sensitive, display down‐regulation of pluripotency factors and induction of mesodermal differentiation. In contrast, seminoma‐like TCam‐2 cells tolerated higher JQ1 concentrations and were resistant to differentiation. ECs xenografted in vivo showed a reduction in tumour size, proliferation rate and angiogenesis in response to JQ1. Finally, the combination of JQ1 and the histone deacetylase inhibitor romidepsin allowed for lower doses and less frequent application, compared with monotherapy. Thus, we propose that JQ1 in combination with romidepsin may serve as a novel therapeutic option for (mixed) TGCTs.
Collapse
Affiliation(s)
- Sina Jostes
- Institute of Pathology, Department of Developmental Pathology, University Medical School, Bonn, Germany
| | - Daniel Nettersheim
- Institute of Pathology, Department of Developmental Pathology, University Medical School, Bonn, Germany
| | - Martin Fellermeyer
- Institute of Pathology, Department of Developmental Pathology, University Medical School, Bonn, Germany
| | - Simon Schneider
- Institute of Pathology, Department of Developmental Pathology, University Medical School, Bonn, Germany
| | - François Hafezi
- Institute of Pathology, Department of Developmental Pathology, University Medical School, Bonn, Germany
| | | | - Valerie Schumacher
- Department of Urology, Boston Children's Hospital, Boston, MA, USA.,Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Matthias Geyer
- Institute of Innate Immunity, Department of Structural Immunology, University Medical School, Bonn, Germany
| | - Glen Kristiansen
- Institute of Pathology, University Medical School, Bonn, Germany
| | - Hubert Schorle
- Institute of Pathology, Department of Developmental Pathology, University Medical School, Bonn, Germany
| |
Collapse
|