Get 20M+ Full-Text Papers For Less Than $1.50/day. Start a 14-Day Trial for You or Your Team.

Learn More →

Renal Medullary Carcinoma: Case Report of an Aggressive Malignancy with Near-Complete Response to Dose-Dense Methotrexate, Vinblastine, Doxorubicin, and Cisplatin Chemotherapy

Renal Medullary Carcinoma: Case Report of an Aggressive Malignancy with Near-Complete Response to... Renal Medullary Carcinoma: Case Report of an Aggressive Malignancy with Near-Complete Response to Dose-Dense Methotrexate, Vinblastine, Doxorubicin, and Cisplatin Chemotherapy div.banner_title_bkg div.trangle { border-color: #172e0d transparent transparent transparent; opacity:0.7; /*new styles start*/ -ms-filter:"progid:DXImageTransform.Microsoft.Alpha(Opacity=70)" ;filter: alpha(opacity=70); /*new styles end*/ } div.banner_title_bkg_if div.trangle { border-color: transparent transparent #172e0d transparent ; opacity:0.7; /*new styles start*/ -ms-filter:"progid:DXImageTransform.Microsoft.Alpha(Opacity=70)" ;filter: alpha(opacity=70); /*new styles end*/ } div.banner_title_bkg div.trangle { width: 348px; } #banner { background-image: url('http://images.hindawi.com/journals/crionm/crionm.banner.jpg'); background-position: 50% 0;} Hindawi Publishing Corporation Home Journals About Us Case Reports in Oncological Medicine About this Journal Submit a Manuscript Table of Contents Journal Menu About this Journal · Abstracting and Indexing · Aims and Scope · Article Processing Charges · Articles in Press · Author Guidelines · Bibliographic Information · Citations to this Journal · Contact Information · Editorial Board · Editorial Workflow · Free eTOC Alerts · Publication Ethics · Reviewers Acknowledgment · Submit a Manuscript · Subscription Information · Table of Contents Abstract Full-Text PDF Full-Text HTML Full-Text ePUB Linked References How to Cite this Article Case Reports in Oncological Medicine Volume 2014 (2014), Article ID 615895, 5 pages http://dx.doi.org/10.1155/2014/615895 Case Report Renal Medullary Carcinoma: Case Report of an Aggressive Malignancy with Near-Complete Response to Dose-Dense Methotrexate, Vinblastine, Doxorubicin, and Cisplatin Chemotherapy Ali Imran Amjad , 1 Hira Ali , 2 Leonard J. Appleman , 1 Jodi Maranchie , 3 Stephen Jackman , 3 Anil Parwani , 4 Rajiv Dhir , 4 Somak Roy , 4 and Rahul A. Parikh 1 1 Division of Hematology and Oncology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA 2 Division of General Internal Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA 3 Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15232, USA 4 Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA Received 16 June 2014; Accepted 5 August 2014; Published 19 August 2014 Academic Editor: Jose I. Mayordomo Copyright © 2014 Ali Imran Amjad et al. This is an open access article distributed under the Creative Commons Attribution License , which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Renal medullary carcinoma (RMC) is a rare but aggressive malignancy affecting young individuals with sickle cell trait. Renal medullary carcinoma commonly presents with advanced or metastatic disease and is associated with a rapidly progressive clinical course and an extremely short overall survival measured in weeks to few months. Due to the rarity of RMC, there is no proven effective therapy and patients are often treated with platinum-based chemotherapy. We report near-complete radiological and pathological response in a patient treated with dose-dense MVAC (methotrexate, vinblastine, doxorubicin, and cisplatin) chemotherapy. The patient underwent consolidation nephrectomy and retroperitoneal lymph node dissection and had a 16-month progression-free survival, one of the longest reported in patients with RMC. 1. Background Renal medullary carcinoma (RMC) was initially described in 1995 in a retrospective study of 34 patients collected over 22 years at the Armed Forces Institute of Pathology. Thirty-three of the 34 patients had sickle cell trait or sickle cell disease; the mean tumor size at diagnosis was 7 cm and median survival following surgery was 12 weeks (range 3–52 weeks) [ 1 ]. In a second smaller retrospective analysis with 6 patients, median age at diagnosis was 24.5 years and time from diagnosis to death was 3 months (range 1–7 months) [ 2 ]. Karyotyping was performed in 4 patients and revealed chromosome 11 monosomy in all 4 patients and chromosome 3 abnormalities in 2 patients [ 2 ]. A retrospective study performed in Brazil, a country with a high incidence of sickle cell trait, identified seven patients with mean age of 22 years (8–69 years) and tumor size ranging from 4 to 12 cm. In this retrospective analysis, survival of patients with advanced RMC ranged from 4 days to 9 months [ 3 ]. A number of other case reports and series have described the role of surgery, chemotherapy, immunotherapy, or radiation in the treatment of RMC and found rapid progression despite these therapies [ 3 – 6 ]. Rathmell and Monk initially reported the use of high-dose intensity MVAC chemotherapy in a case-series of three patients with RMC [ 7 ]. They noted a significant improvement in both palliation and survival with the use of this regimen. In the current paper, we describe a young 23-year-old subject with sickle cell trait, diagnosed with diffusely metastatic RMC. In spite of having advanced disease, she achieved an excellent response to aggressive cytotoxic chemotherapy with a progression-free survival (PFS) of 16 months. 2. Case Presentation A twenty-three-year-old African-American female with sickle cell trait presented to the emergency room with periumbilical and right-sided back pain associated with poor appetite and ten-pound weight loss over 4 months. She denied gross hematuria, dysuria, or additional urinary symptoms. She was a previously healthy nonsmoker with paternal family history of sickle cell trait. Physical examination was pertinent for fullness in the right flank and left supraclavicular lymphadenopathy. Laboratory findings revealed hemoglobin of 11.3 g/dL, platelet count of 269,000 per mm 3 , and normal LDH of 147 IU/L. Computed tomography (CT) scan of the neck, chest, abdomen, and pelvis demonstrated a 12 cm heterogeneous right renal mass (Figure 1(a) ) with retrocaval, aortocaval, and paraaortic lymphadenopathy. In addition, there were bilateral pulmonary nodules (largest in right upper lobe measuring 1.8 × 1.4 cm), left pleural-based nodules (largest at the level of diaphragmatic pleura measuring 2.6 × 1.9 cm), and right hilar (2.5 × 2.2 cm), left supraclavicular, and bilateral cervical lymphadenopathy. MRI of the brain showed no evidence for intracranial metastasis. Figure 1: (a) CT scan of abdomen showing necrotic right-sided tumor replacing the kidney. (b) CT scan after chemotherapy. Biopsy of left supraclavicular lymph node revealed high-grade renal medullary carcinoma with prominent lymphovascular tumor emboli. The tumor cells were eosinophilic, containing large nuclei and focally prominent nucleoli with brisk mitotic activity. The neoplastic cells stained positive for AE1/AE3, cytokeratin CAM 5.2, epithelial membrane antigen (EMA), E-cadherin, cytokeratin 7, and cytokeratin 19 on immunohistochemical analyses (Figure 2 ). The cells stained weakly positive for c-Kit and negative for cytokeratin 20, S100 protein, carbonic anhydrase IX, mucicarmine, and CD10. Cytogenetic studies performed on the lymph node isolated a clone of cells with duplication of the long arm of chromosome 1 (resulting in partial trisomy 1q) and a derivative chromosome 22 with chromatin material of unknown origin attached to the long arm (resulting in partial monosomy 22q). Fluorescence in situ hybridization (FISH) for transcription factor E3 ( TFE ) gene translocation was negative. Figure 2: Supraclavicular lymph node (20x) with immunohistochemistry for CAM 5.2 and EMA. Treatment was initiated with dose-dense (dd) MVAC consisting of methotrexate 30 mg/m 2 on day 1 followed by vinblastine 3 mg/m 2 , doxorubicin 30 mg/m 2 , and cisplatin 70 mg/m 2 on day 2 of a 14-day cycle as described for metastatic urothelial carcinomas [ 8 ]. She received growth factor support with pegfilgastrim on day 3 to reduce the risk of neutropenic fevers. Treatment with four cycles of dd-MVAC was associated with a significant reduction in size of renal mass from 9.3 cm to 5.9 cm with areas of necrosis on a CT scan of chest, abdomen, and pelvis (Figure 1(b) ). There was near-complete resolution of right upper lobe and left lower lobe parenchymal lung nodules and retroperitoneal lymphadenopathy. Multiple pleural-based nodules in the left hemithorax and right hilar lymph node decreased. Two additional cycles of dd-MVAC were administered and a follow-up CT scan showed continued improvement in size of renal mass to 4.4 cm, improvement in retroperitoneal, hilar lymphadenopathy, and parenchymal lung lesions. The subject developed grade I renal insufficiency and grade II chemotherapy-induced nausea and vomiting (CINV) for which the cisplatin dose was reduced by 25% after the initial two cycles. Overall, she tolerated six cycles of dd-MVAC extremely well, with no hospitalizations. After achieving an excellent partial response to neoadjuvant chemotherapy, the patient underwent right radical nephrectomy with complete, bilateral retroperitoneal lymph node dissection without complications. Pathology revealed a gross tumor resection size of 6.2 cm. Viable carcinoma comprised approximately 2–4% of the tumor volume showing neoplastic cells with rhabdoid features in a background of extensive fibrosis and chronic interstitial inflammation. All surgical margins were negative for tumor. Two of 11 lymph nodes (one paracaval and one preaortic from the level of the aortic bifurcation) were involved with carcinoma (0.7 cm in greatest dimension) with no extracapsular extension. Pathologic TNM stage was assessed as pT1b N1 M1. Immunohistochemistry revealed a complete loss of SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily B member 1 (SMARCB1) expression. Subsequently, she underwent video assisted thoracoscopy and wedge resection of right lower lung lobe. Pathology showed inflammatory changes with no evidence of residual malignancy. Follow-up imaging in the form of CT chest, abdomen, and pelvis was obtained every four months and at one year from diagnosis she showed no recurrence of disease. At 16 months from initial diagnosis, she developed shortness of breath and restaging scans showed disease recurrence with hepatic, pulmonary lesions and bilateral pleural effusions. She had a rapidly progressive decline after relapse and died without receiving any further therapy. 3. Discussion Renal medullary carcinoma (RMC) is a rare form of aggressive kidney cancer (comprising <1% of all kidney cancers) and is exclusively restricted to patients with sickle cell trait or disease. It typically presents in young patients (male : female ratio 2 : 1) and follows an extremely aggressive clinical course with a poor outcome. We report a patient presenting with widely metastatic disease, with bulky renal primary and involvement of lung, pleura, and mediastinal and retroperitoneal lymph nodes. The patient had an excellent response to dose-dense MVAC regimen (methotrexate, vinblastine, doxorubicin, and cisplatin). After completion of six cycles of chemotherapy there was near-complete radiographic response and pathologic response at the primary 12 cm renal mass and surrounding lymph nodes. The patient eventually developed hepatic and pulmonary disease recurrence with a progression-free survival of 16 months. Patients with RMC have an aggressive course and with cytotoxic regimens, survival is only weeks to a few months. Most patients respond to initial platinum-based chemotherapy but responses are rarely durable. The mechanism of this acquired resistance to cytotoxic chemotherapy is not clearly understood. The MVAC chemotherapy regimen, approved for use in advanced or metastatic transitional cell carcinoma of urinary bladder, has been previously utilized in RMC. Rathmell and Monk treated three patients with dose-dense MVAC and demonstrated partial responses with one patient achieving a 90% response by RECIST criteria after six cycles of chemotherapy. That patient relapsed and died at 12 months from initial diagnosis [ 7 ]. Walsh and colleagues reported a complete pathologic response to a combination of paclitaxel, gemcitabine with carboplatin in an 11-year-old patient with diffusely metastatic disease (lung, bone, and liver) [ 13 ]. This patient was diagnosed with left supraclavicular node biopsy and underwent neoadjuvant chemotherapy prior to resection of primary tumor and pulmonary metastasis. However, the patient had early relapse with leptomeningeal disease and died 24 months after initial diagnosis [ 13 ]. Very few other case reports of complete radiological responses have been described (Table 1 ). Pirich and colleagues initially reported a 12-year-old with sickle cell trait with a 6-month PFS to MVAC and an overall survival of 15 months [ 14 ]. Table 1: Case reports of complete radiological responses, regimens used, and survival of patients with renal medullary carcinoma. A number of genetic abnormalities with potential pathophysiologic or therapeutic implications have been recently described in RMC. Translocation t (9; 22) involving bcr-abl has been described; however use of imatinib has not yielded any meaningful responses [ 9 , 10 ]. Rearrangement of the ALK gene with translocation of the ALK receptor tyrosine kinase to a cytoskeleton protein vinculin has been described in a t (2; 10) bearing RMC [ 15 ]. In a recent study, RMC tumor specimens were noted to have an absence of SMARCB1/INI1 by immunohistochemistry. This correlates with loss of heterozygosity at the SMARCB1/INI1 gene locus on chromosome 22 [ 16 , 17 ]. Of note, our patient also demonstrated a loss of SMARCB1/INI1 expression by immunohistochemistry, with karyotyping revealing partial monosomy of chromosome 22q. SMARCB1 is a tumor suppressor gene, which plays a role in chromatin remodeling, cell cycle control, and regulation of cytoskeletal dynamics [ 18 – 20 ]. It is not entirely clear how this gene may play a role in the pathogenesis of RMC. Prior reports have noted increased expression of hypoxia inducible factor (HIF), vascular endothelial growth factor (VEGF), and TP53 in RMC [ 21 ]. Limited data on gene expression profiling of RMC using cDNA has shown resemblance to transitional cell carcinoma of the urinary bladder, which is supported with the rationale of treating these patients with MVAC, a regimen utilized commonly in transitional cell carcinomas of the bladder [ 22 ]. Whole-genome expression analysis in a patient who had complete remission for 9 months after second-line doxorubicin-based therapy showed high expression of topoisomerase II [ 23 ]. This expression profile may explain the unusual chemosensitivity of this patient to topoisomerase II inhibitor-based therapy. In the era of next generation sequencing, it is worth exploring whether sequencing this rare form of cancer will identify any driver-mutations, which can be targeted for therapeutic benefit. 4. Conclusion In summary, we observed an excellent clinical and radiologic response in our patient with diffusely metastatic RMC with a PFS of 16 months on treatment with dd-MVAC followed by consolidation surgery. Thus, aggressive intervention of platinum-based chemotherapy can achieve prolonged responses with significant palliation of symptoms and should be strongly considered. Abbreviations RMC: Renal medullary carcinoma LDH: Lactate dehydrogenase EMA: Epithelial membrane antigen FISH: Fluorescence in situ hybridization MVAC: Methotrexate, vinblastine, doxorubicin, and cisplatin SMARCB1/INI-1: SWI/SNF related, matrix-associated, actin-dependent regulator of chromatin, subfamily b, member 1 RECIST: Response evaluation criteria in solid tumors. Consent Written informed consent was obtained from the patient’s spouse for publication of this case report and the accompanying figures. A copy of the written consent is available for review. Conflict of Interests The authors declare no conflict of interests. References C. J. Davis Jr., F. K. Mostofi, and I. A. Sesterhenn, “Renal medullary carcinoma: the seventh sickle cell nephropathy,” American Journal of Surgical Pathology , vol. 19, no. 1, pp. 1–11, 1995. View at Publisher · View at Google Scholar · View at Scopus R. A. Avery, J. E. Harris, C. J. Davis Jr., D. S. Borgaonkar, J. C. Byrd, and R. B. Weiss, “Renal medullary carcinoma: clinical and therapeutic aspects of a newly described tumor,” Cancer , vol. 78, no. 1, pp. 128–132, 1996. I. C. Watanabe, A. Billis, M. S. Guimarães et al., “Renal medullary carcinoma: report of seven cases from Brazil,” Modern Pathology , vol. 20, no. 9, pp. 914–920, 2007. View at Publisher · View at Google Scholar · View at Scopus C. L. Coogan, C. F. McKiel Jr., M. J. Flanagan, T. P. Bormes, and T. G. Matkov, “Renal medullary carcinoma in patients with sickle cell trait,” Urology , vol. 51, no. 6, pp. 1049–1050, 1998. View at Publisher · View at Google Scholar · View at Scopus W. G. Noguera-Irizarry, H. Hibshoosh, and K. P. Papadopoulos, “Renal medullary carcinoma: case report and review of the literature,” The American Journal of Clinical Oncology: Cancer Clinical Trials , vol. 26, no. 5, pp. 489–492, 2003. View at Publisher · View at Google Scholar · View at Scopus A. A. Hakimi, P. T. Koi, P. M. Milhoua et al., “Renal medullary carcinoma: the Bronx experience,” Urology , vol. 70, no. 5, pp. 878–882, 2007. View at Publisher · View at Google Scholar · View at Scopus W. K. Rathmell and J. P. Monk, “High-dose-intensity mvac for advanced renal medullary carcinoma: report of three cases and literature review,” Urology , vol. 72, no. 3, pp. 659–663, 2008. View at Publisher · View at Google Scholar · View at Scopus C. N. Sternberg, P. H. M. de Mulder, J. H. Schornagel et al., “Randomized phase III trial of high-dose-intensity methotrexate, vinblastine, doxorubicin, and cisplatin (MVAC) chemotherapy and recombinant human granulocyte colony-stimulating factor versus classic MVAC in advanced urothelial tract tumors: european organization for research and treatment of cancer protocol no. 30924,” Journal of Clinical Oncology , vol. 19, no. 10, pp. 2638–2646, 2001. View at Scopus J. Stahlschmidt, C. Cullinane, P. Roberts, and S. V. Picton, “Renal medullary carcinoma: prolonged remission with chemotherapy, immunohistochemical characterisation and evidence of bcr/abl rearrangement,” Medical and Pediatric Oncology , vol. 33, no. 6, pp. 551–557, 1999. L. Simpson, X. He, M. Pins et al., “Renal medullary carcinoma and ABL gene amplification,” Journal of Urology , vol. 173, no. 6, pp. 1883–1888, 2005. View at Publisher · View at Google Scholar · View at Scopus J. J. Strouse, M. Spevak, A. K. Mack, R. J. Arceci, D. Small, and D. M. Loeb, “Significant responses to platinum-based chemotherapy in renal medullary carcinoma,” Pediatric Blood & Cancer , vol. 44, no. 4, pp. 407–411, 2005. View at Publisher · View at Google Scholar · View at Scopus E. A. Ronnen, G. V. Kondagunta, and R. J. Motzer, “Medullary renal cell carcinoma and response to therapy with bortezomib,” Journal of Clinical Oncology , vol. 24, no. 9, article e14, 2006. View at Publisher · View at Google Scholar · View at Scopus A. Walsh, D. R. Kelly, Y. N. Vaid, L. M. Hilliard, and G. K. Friedman, “Complete response to carboplatin, gemcitabine, and paclitaxel in a patient with advanced metastatic renal medullary carcinoma,” Pediatric Blood and Cancer , vol. 55, no. 6, pp. 1217–1220, 2010. View at Publisher · View at Google Scholar · View at Scopus L. M. Pirich, P. Chou, and D. O. Walterhouse, “Prolonged survival of a patient with sickle cell trait and metastatic renal medullary carcinoma,” Journal of Pediatric Hematology/Oncology , vol. 21, no. 1, pp. 67–69, 1999. View at Publisher · View at Google Scholar · View at Scopus A. Mariño-Enríquez, W. Ou, C. B. Weldon, J. A. Fletcher, and A. R. Pérez-Atayde, “ALK rearrangement in sickle cell trait-associated renal medullary carcinoma,” Genes Chromosomes and Cancer , vol. 50, no. 3, pp. 146–153, 2011. View at Publisher · View at Google Scholar · View at Scopus Q. Liu, S. Galli, R. Srinivasan, W. M. Linehan, M. Tsokos, and M. J. Merino, “Renal medullary carcinoma: molecular, immunohistochemistry, and morphologic correlation,” The American Journal of Surgical Pathology , vol. 37, no. 3, pp. 368–374, 2013. View at Publisher · View at Google Scholar · View at Scopus J. Calderaro, J. Moroch, G. Pierron et al., “SMARCB1/INI1 inactivation in renal medullary carcinoma,” Histopathology , vol. 61, no. 3, pp. 428–435, 2012. View at Publisher · View at Google Scholar · View at Scopus A. Klochendler-Yeivin, E. Picarsky, and M. Yaniv, “Increased DNA damage sensitivity and apoptosis in cells lacking the Snf5/Ini1 subunit of the SWI/SNF chromatin remodeling complex,” Molecular and Cellular Biology , vol. 26, no. 7, pp. 2661–2674, 2006. View at Publisher · View at Google Scholar · View at Scopus S. Medjkane, E. Novikov, I. Versteege, and O. Delattre, “The tumor suppressor hSNF5/INI1 modulates cell growth and actin cytoskeleton organization,” Cancer Research , vol. 64, no. 10, pp. 3406–3413, 2004. View at Publisher · View at Google Scholar · View at Scopus I. Versteege, S. Medjkane, D. Rouillard, and O. Delattre, “A key role of the hSNF5/INI1 tumour suppressor in the control of the G1-S transition of the cell cycle,” Oncogene , vol. 21, no. 42, pp. 6403–6412, 2002. View at Publisher · View at Google Scholar · View at Scopus M. A. Swartz, J. Karth, D. T. Schneider, R. Rodriguez, J. B. Beckwith, and E. J. Perlman, “Renal medullary carcinoma: clinical, pathologic, immunohistochemical, and genetic analysis with pathogenetic implications,” Urology , vol. 60, no. 6, pp. 1083–1089, 2002. View at Publisher · View at Google Scholar · View at Scopus X. J. Yang, J. Sugimura, M. S. Tretiakova et al., “Gene expression profiling of renal medullary carcinoma: potential clinical relevance,” Cancer , vol. 100, no. 5, pp. 976–985, 2004. View at Publisher · View at Google Scholar · View at Scopus E. M. Schaeffer, T. J. Guzzo, K. A. Furge et al., “Renal medullary carcinoma: Molecular, pathological and clinical evidence for treatment with topoisomerase-inhibiting therapy,” BJU International , vol. 106, no. 1, pp. 62–65, 2010. View at Publisher · View at Google Scholar · View at Scopus var _gaq = _gaq || []; _gaq.push(['_setAccount', 'UA-8578054-2']); _gaq.push(['_trackPageview']); (function () { var ga = document.createElement('script'); ga.type = 'text/javascript'; ga.async = true; ga.src = ('https:' == document.location.protocol ? 'https://ssl' : 'http://www') + '.google-analytics.com/ga.js'; var s = document.getElementsByTagName('script')[0]; s.parentNode.insertBefore(ga, s); })(); http://www.deepdyve.com/assets/images/DeepDyve-Logo-lg.png Case Reports in Oncological Medicine Hindawi Publishing Corporation

Renal Medullary Carcinoma: Case Report of an Aggressive Malignancy with Near-Complete Response to Dose-Dense Methotrexate, Vinblastine, Doxorubicin, and Cisplatin Chemotherapy

Loading next page...
 
/lp/hindawi-publishing-corporation/renal-medullary-carcinoma-case-report-of-an-aggressive-malignancy-with-Ch2TX8itNS

References

References for this paper are not available at this time. We will be adding them shortly, thank you for your patience.

Publisher
Hindawi Publishing Corporation
Copyright
Copyright © 2014 Ali Imran Amjad et al.
ISSN
2090-6706
eISSN
2090-6714
Publisher site
See Article on Publisher Site

Abstract

Renal Medullary Carcinoma: Case Report of an Aggressive Malignancy with Near-Complete Response to Dose-Dense Methotrexate, Vinblastine, Doxorubicin, and Cisplatin Chemotherapy div.banner_title_bkg div.trangle { border-color: #172e0d transparent transparent transparent; opacity:0.7; /*new styles start*/ -ms-filter:"progid:DXImageTransform.Microsoft.Alpha(Opacity=70)" ;filter: alpha(opacity=70); /*new styles end*/ } div.banner_title_bkg_if div.trangle { border-color: transparent transparent #172e0d transparent ; opacity:0.7; /*new styles start*/ -ms-filter:"progid:DXImageTransform.Microsoft.Alpha(Opacity=70)" ;filter: alpha(opacity=70); /*new styles end*/ } div.banner_title_bkg div.trangle { width: 348px; } #banner { background-image: url('http://images.hindawi.com/journals/crionm/crionm.banner.jpg'); background-position: 50% 0;} Hindawi Publishing Corporation Home Journals About Us Case Reports in Oncological Medicine About this Journal Submit a Manuscript Table of Contents Journal Menu About this Journal · Abstracting and Indexing · Aims and Scope · Article Processing Charges · Articles in Press · Author Guidelines · Bibliographic Information · Citations to this Journal · Contact Information · Editorial Board · Editorial Workflow · Free eTOC Alerts · Publication Ethics · Reviewers Acknowledgment · Submit a Manuscript · Subscription Information · Table of Contents Abstract Full-Text PDF Full-Text HTML Full-Text ePUB Linked References How to Cite this Article Case Reports in Oncological Medicine Volume 2014 (2014), Article ID 615895, 5 pages http://dx.doi.org/10.1155/2014/615895 Case Report Renal Medullary Carcinoma: Case Report of an Aggressive Malignancy with Near-Complete Response to Dose-Dense Methotrexate, Vinblastine, Doxorubicin, and Cisplatin Chemotherapy Ali Imran Amjad , 1 Hira Ali , 2 Leonard J. Appleman , 1 Jodi Maranchie , 3 Stephen Jackman , 3 Anil Parwani , 4 Rajiv Dhir , 4 Somak Roy , 4 and Rahul A. Parikh 1 1 Division of Hematology and Oncology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA 2 Division of General Internal Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA 3 Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15232, USA 4 Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA Received 16 June 2014; Accepted 5 August 2014; Published 19 August 2014 Academic Editor: Jose I. Mayordomo Copyright © 2014 Ali Imran Amjad et al. This is an open access article distributed under the Creative Commons Attribution License , which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Renal medullary carcinoma (RMC) is a rare but aggressive malignancy affecting young individuals with sickle cell trait. Renal medullary carcinoma commonly presents with advanced or metastatic disease and is associated with a rapidly progressive clinical course and an extremely short overall survival measured in weeks to few months. Due to the rarity of RMC, there is no proven effective therapy and patients are often treated with platinum-based chemotherapy. We report near-complete radiological and pathological response in a patient treated with dose-dense MVAC (methotrexate, vinblastine, doxorubicin, and cisplatin) chemotherapy. The patient underwent consolidation nephrectomy and retroperitoneal lymph node dissection and had a 16-month progression-free survival, one of the longest reported in patients with RMC. 1. Background Renal medullary carcinoma (RMC) was initially described in 1995 in a retrospective study of 34 patients collected over 22 years at the Armed Forces Institute of Pathology. Thirty-three of the 34 patients had sickle cell trait or sickle cell disease; the mean tumor size at diagnosis was 7 cm and median survival following surgery was 12 weeks (range 3–52 weeks) [ 1 ]. In a second smaller retrospective analysis with 6 patients, median age at diagnosis was 24.5 years and time from diagnosis to death was 3 months (range 1–7 months) [ 2 ]. Karyotyping was performed in 4 patients and revealed chromosome 11 monosomy in all 4 patients and chromosome 3 abnormalities in 2 patients [ 2 ]. A retrospective study performed in Brazil, a country with a high incidence of sickle cell trait, identified seven patients with mean age of 22 years (8–69 years) and tumor size ranging from 4 to 12 cm. In this retrospective analysis, survival of patients with advanced RMC ranged from 4 days to 9 months [ 3 ]. A number of other case reports and series have described the role of surgery, chemotherapy, immunotherapy, or radiation in the treatment of RMC and found rapid progression despite these therapies [ 3 – 6 ]. Rathmell and Monk initially reported the use of high-dose intensity MVAC chemotherapy in a case-series of three patients with RMC [ 7 ]. They noted a significant improvement in both palliation and survival with the use of this regimen. In the current paper, we describe a young 23-year-old subject with sickle cell trait, diagnosed with diffusely metastatic RMC. In spite of having advanced disease, she achieved an excellent response to aggressive cytotoxic chemotherapy with a progression-free survival (PFS) of 16 months. 2. Case Presentation A twenty-three-year-old African-American female with sickle cell trait presented to the emergency room with periumbilical and right-sided back pain associated with poor appetite and ten-pound weight loss over 4 months. She denied gross hematuria, dysuria, or additional urinary symptoms. She was a previously healthy nonsmoker with paternal family history of sickle cell trait. Physical examination was pertinent for fullness in the right flank and left supraclavicular lymphadenopathy. Laboratory findings revealed hemoglobin of 11.3 g/dL, platelet count of 269,000 per mm 3 , and normal LDH of 147 IU/L. Computed tomography (CT) scan of the neck, chest, abdomen, and pelvis demonstrated a 12 cm heterogeneous right renal mass (Figure 1(a) ) with retrocaval, aortocaval, and paraaortic lymphadenopathy. In addition, there were bilateral pulmonary nodules (largest in right upper lobe measuring 1.8 × 1.4 cm), left pleural-based nodules (largest at the level of diaphragmatic pleura measuring 2.6 × 1.9 cm), and right hilar (2.5 × 2.2 cm), left supraclavicular, and bilateral cervical lymphadenopathy. MRI of the brain showed no evidence for intracranial metastasis. Figure 1: (a) CT scan of abdomen showing necrotic right-sided tumor replacing the kidney. (b) CT scan after chemotherapy. Biopsy of left supraclavicular lymph node revealed high-grade renal medullary carcinoma with prominent lymphovascular tumor emboli. The tumor cells were eosinophilic, containing large nuclei and focally prominent nucleoli with brisk mitotic activity. The neoplastic cells stained positive for AE1/AE3, cytokeratin CAM 5.2, epithelial membrane antigen (EMA), E-cadherin, cytokeratin 7, and cytokeratin 19 on immunohistochemical analyses (Figure 2 ). The cells stained weakly positive for c-Kit and negative for cytokeratin 20, S100 protein, carbonic anhydrase IX, mucicarmine, and CD10. Cytogenetic studies performed on the lymph node isolated a clone of cells with duplication of the long arm of chromosome 1 (resulting in partial trisomy 1q) and a derivative chromosome 22 with chromatin material of unknown origin attached to the long arm (resulting in partial monosomy 22q). Fluorescence in situ hybridization (FISH) for transcription factor E3 ( TFE ) gene translocation was negative. Figure 2: Supraclavicular lymph node (20x) with immunohistochemistry for CAM 5.2 and EMA. Treatment was initiated with dose-dense (dd) MVAC consisting of methotrexate 30 mg/m 2 on day 1 followed by vinblastine 3 mg/m 2 , doxorubicin 30 mg/m 2 , and cisplatin 70 mg/m 2 on day 2 of a 14-day cycle as described for metastatic urothelial carcinomas [ 8 ]. She received growth factor support with pegfilgastrim on day 3 to reduce the risk of neutropenic fevers. Treatment with four cycles of dd-MVAC was associated with a significant reduction in size of renal mass from 9.3 cm to 5.9 cm with areas of necrosis on a CT scan of chest, abdomen, and pelvis (Figure 1(b) ). There was near-complete resolution of right upper lobe and left lower lobe parenchymal lung nodules and retroperitoneal lymphadenopathy. Multiple pleural-based nodules in the left hemithorax and right hilar lymph node decreased. Two additional cycles of dd-MVAC were administered and a follow-up CT scan showed continued improvement in size of renal mass to 4.4 cm, improvement in retroperitoneal, hilar lymphadenopathy, and parenchymal lung lesions. The subject developed grade I renal insufficiency and grade II chemotherapy-induced nausea and vomiting (CINV) for which the cisplatin dose was reduced by 25% after the initial two cycles. Overall, she tolerated six cycles of dd-MVAC extremely well, with no hospitalizations. After achieving an excellent partial response to neoadjuvant chemotherapy, the patient underwent right radical nephrectomy with complete, bilateral retroperitoneal lymph node dissection without complications. Pathology revealed a gross tumor resection size of 6.2 cm. Viable carcinoma comprised approximately 2–4% of the tumor volume showing neoplastic cells with rhabdoid features in a background of extensive fibrosis and chronic interstitial inflammation. All surgical margins were negative for tumor. Two of 11 lymph nodes (one paracaval and one preaortic from the level of the aortic bifurcation) were involved with carcinoma (0.7 cm in greatest dimension) with no extracapsular extension. Pathologic TNM stage was assessed as pT1b N1 M1. Immunohistochemistry revealed a complete loss of SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily B member 1 (SMARCB1) expression. Subsequently, she underwent video assisted thoracoscopy and wedge resection of right lower lung lobe. Pathology showed inflammatory changes with no evidence of residual malignancy. Follow-up imaging in the form of CT chest, abdomen, and pelvis was obtained every four months and at one year from diagnosis she showed no recurrence of disease. At 16 months from initial diagnosis, she developed shortness of breath and restaging scans showed disease recurrence with hepatic, pulmonary lesions and bilateral pleural effusions. She had a rapidly progressive decline after relapse and died without receiving any further therapy. 3. Discussion Renal medullary carcinoma (RMC) is a rare form of aggressive kidney cancer (comprising <1% of all kidney cancers) and is exclusively restricted to patients with sickle cell trait or disease. It typically presents in young patients (male : female ratio 2 : 1) and follows an extremely aggressive clinical course with a poor outcome. We report a patient presenting with widely metastatic disease, with bulky renal primary and involvement of lung, pleura, and mediastinal and retroperitoneal lymph nodes. The patient had an excellent response to dose-dense MVAC regimen (methotrexate, vinblastine, doxorubicin, and cisplatin). After completion of six cycles of chemotherapy there was near-complete radiographic response and pathologic response at the primary 12 cm renal mass and surrounding lymph nodes. The patient eventually developed hepatic and pulmonary disease recurrence with a progression-free survival of 16 months. Patients with RMC have an aggressive course and with cytotoxic regimens, survival is only weeks to a few months. Most patients respond to initial platinum-based chemotherapy but responses are rarely durable. The mechanism of this acquired resistance to cytotoxic chemotherapy is not clearly understood. The MVAC chemotherapy regimen, approved for use in advanced or metastatic transitional cell carcinoma of urinary bladder, has been previously utilized in RMC. Rathmell and Monk treated three patients with dose-dense MVAC and demonstrated partial responses with one patient achieving a 90% response by RECIST criteria after six cycles of chemotherapy. That patient relapsed and died at 12 months from initial diagnosis [ 7 ]. Walsh and colleagues reported a complete pathologic response to a combination of paclitaxel, gemcitabine with carboplatin in an 11-year-old patient with diffusely metastatic disease (lung, bone, and liver) [ 13 ]. This patient was diagnosed with left supraclavicular node biopsy and underwent neoadjuvant chemotherapy prior to resection of primary tumor and pulmonary metastasis. However, the patient had early relapse with leptomeningeal disease and died 24 months after initial diagnosis [ 13 ]. Very few other case reports of complete radiological responses have been described (Table 1 ). Pirich and colleagues initially reported a 12-year-old with sickle cell trait with a 6-month PFS to MVAC and an overall survival of 15 months [ 14 ]. Table 1: Case reports of complete radiological responses, regimens used, and survival of patients with renal medullary carcinoma. A number of genetic abnormalities with potential pathophysiologic or therapeutic implications have been recently described in RMC. Translocation t (9; 22) involving bcr-abl has been described; however use of imatinib has not yielded any meaningful responses [ 9 , 10 ]. Rearrangement of the ALK gene with translocation of the ALK receptor tyrosine kinase to a cytoskeleton protein vinculin has been described in a t (2; 10) bearing RMC [ 15 ]. In a recent study, RMC tumor specimens were noted to have an absence of SMARCB1/INI1 by immunohistochemistry. This correlates with loss of heterozygosity at the SMARCB1/INI1 gene locus on chromosome 22 [ 16 , 17 ]. Of note, our patient also demonstrated a loss of SMARCB1/INI1 expression by immunohistochemistry, with karyotyping revealing partial monosomy of chromosome 22q. SMARCB1 is a tumor suppressor gene, which plays a role in chromatin remodeling, cell cycle control, and regulation of cytoskeletal dynamics [ 18 – 20 ]. It is not entirely clear how this gene may play a role in the pathogenesis of RMC. Prior reports have noted increased expression of hypoxia inducible factor (HIF), vascular endothelial growth factor (VEGF), and TP53 in RMC [ 21 ]. Limited data on gene expression profiling of RMC using cDNA has shown resemblance to transitional cell carcinoma of the urinary bladder, which is supported with the rationale of treating these patients with MVAC, a regimen utilized commonly in transitional cell carcinomas of the bladder [ 22 ]. Whole-genome expression analysis in a patient who had complete remission for 9 months after second-line doxorubicin-based therapy showed high expression of topoisomerase II [ 23 ]. This expression profile may explain the unusual chemosensitivity of this patient to topoisomerase II inhibitor-based therapy. In the era of next generation sequencing, it is worth exploring whether sequencing this rare form of cancer will identify any driver-mutations, which can be targeted for therapeutic benefit. 4. Conclusion In summary, we observed an excellent clinical and radiologic response in our patient with diffusely metastatic RMC with a PFS of 16 months on treatment with dd-MVAC followed by consolidation surgery. Thus, aggressive intervention of platinum-based chemotherapy can achieve prolonged responses with significant palliation of symptoms and should be strongly considered. Abbreviations RMC: Renal medullary carcinoma LDH: Lactate dehydrogenase EMA: Epithelial membrane antigen FISH: Fluorescence in situ hybridization MVAC: Methotrexate, vinblastine, doxorubicin, and cisplatin SMARCB1/INI-1: SWI/SNF related, matrix-associated, actin-dependent regulator of chromatin, subfamily b, member 1 RECIST: Response evaluation criteria in solid tumors. Consent Written informed consent was obtained from the patient’s spouse for publication of this case report and the accompanying figures. A copy of the written consent is available for review. Conflict of Interests The authors declare no conflict of interests. References C. J. Davis Jr., F. K. Mostofi, and I. A. Sesterhenn, “Renal medullary carcinoma: the seventh sickle cell nephropathy,” American Journal of Surgical Pathology , vol. 19, no. 1, pp. 1–11, 1995. View at Publisher · View at Google Scholar · View at Scopus R. A. Avery, J. E. Harris, C. J. Davis Jr., D. S. Borgaonkar, J. C. Byrd, and R. B. Weiss, “Renal medullary carcinoma: clinical and therapeutic aspects of a newly described tumor,” Cancer , vol. 78, no. 1, pp. 128–132, 1996. I. C. Watanabe, A. Billis, M. S. Guimarães et al., “Renal medullary carcinoma: report of seven cases from Brazil,” Modern Pathology , vol. 20, no. 9, pp. 914–920, 2007. View at Publisher · View at Google Scholar · View at Scopus C. L. Coogan, C. F. McKiel Jr., M. J. Flanagan, T. P. Bormes, and T. G. Matkov, “Renal medullary carcinoma in patients with sickle cell trait,” Urology , vol. 51, no. 6, pp. 1049–1050, 1998. View at Publisher · View at Google Scholar · View at Scopus W. G. Noguera-Irizarry, H. Hibshoosh, and K. P. Papadopoulos, “Renal medullary carcinoma: case report and review of the literature,” The American Journal of Clinical Oncology: Cancer Clinical Trials , vol. 26, no. 5, pp. 489–492, 2003. View at Publisher · View at Google Scholar · View at Scopus A. A. Hakimi, P. T. Koi, P. M. Milhoua et al., “Renal medullary carcinoma: the Bronx experience,” Urology , vol. 70, no. 5, pp. 878–882, 2007. View at Publisher · View at Google Scholar · View at Scopus W. K. Rathmell and J. P. Monk, “High-dose-intensity mvac for advanced renal medullary carcinoma: report of three cases and literature review,” Urology , vol. 72, no. 3, pp. 659–663, 2008. View at Publisher · View at Google Scholar · View at Scopus C. N. Sternberg, P. H. M. de Mulder, J. H. Schornagel et al., “Randomized phase III trial of high-dose-intensity methotrexate, vinblastine, doxorubicin, and cisplatin (MVAC) chemotherapy and recombinant human granulocyte colony-stimulating factor versus classic MVAC in advanced urothelial tract tumors: european organization for research and treatment of cancer protocol no. 30924,” Journal of Clinical Oncology , vol. 19, no. 10, pp. 2638–2646, 2001. View at Scopus J. Stahlschmidt, C. Cullinane, P. Roberts, and S. V. Picton, “Renal medullary carcinoma: prolonged remission with chemotherapy, immunohistochemical characterisation and evidence of bcr/abl rearrangement,” Medical and Pediatric Oncology , vol. 33, no. 6, pp. 551–557, 1999. L. Simpson, X. He, M. Pins et al., “Renal medullary carcinoma and ABL gene amplification,” Journal of Urology , vol. 173, no. 6, pp. 1883–1888, 2005. View at Publisher · View at Google Scholar · View at Scopus J. J. Strouse, M. Spevak, A. K. Mack, R. J. Arceci, D. Small, and D. M. Loeb, “Significant responses to platinum-based chemotherapy in renal medullary carcinoma,” Pediatric Blood & Cancer , vol. 44, no. 4, pp. 407–411, 2005. View at Publisher · View at Google Scholar · View at Scopus E. A. Ronnen, G. V. Kondagunta, and R. J. Motzer, “Medullary renal cell carcinoma and response to therapy with bortezomib,” Journal of Clinical Oncology , vol. 24, no. 9, article e14, 2006. View at Publisher · View at Google Scholar · View at Scopus A. Walsh, D. R. Kelly, Y. N. Vaid, L. M. Hilliard, and G. K. Friedman, “Complete response to carboplatin, gemcitabine, and paclitaxel in a patient with advanced metastatic renal medullary carcinoma,” Pediatric Blood and Cancer , vol. 55, no. 6, pp. 1217–1220, 2010. View at Publisher · View at Google Scholar · View at Scopus L. M. Pirich, P. Chou, and D. O. Walterhouse, “Prolonged survival of a patient with sickle cell trait and metastatic renal medullary carcinoma,” Journal of Pediatric Hematology/Oncology , vol. 21, no. 1, pp. 67–69, 1999. View at Publisher · View at Google Scholar · View at Scopus A. Mariño-Enríquez, W. Ou, C. B. Weldon, J. A. Fletcher, and A. R. Pérez-Atayde, “ALK rearrangement in sickle cell trait-associated renal medullary carcinoma,” Genes Chromosomes and Cancer , vol. 50, no. 3, pp. 146–153, 2011. View at Publisher · View at Google Scholar · View at Scopus Q. Liu, S. Galli, R. Srinivasan, W. M. Linehan, M. Tsokos, and M. J. Merino, “Renal medullary carcinoma: molecular, immunohistochemistry, and morphologic correlation,” The American Journal of Surgical Pathology , vol. 37, no. 3, pp. 368–374, 2013. View at Publisher · View at Google Scholar · View at Scopus J. Calderaro, J. Moroch, G. Pierron et al., “SMARCB1/INI1 inactivation in renal medullary carcinoma,” Histopathology , vol. 61, no. 3, pp. 428–435, 2012. View at Publisher · View at Google Scholar · View at Scopus A. Klochendler-Yeivin, E. Picarsky, and M. Yaniv, “Increased DNA damage sensitivity and apoptosis in cells lacking the Snf5/Ini1 subunit of the SWI/SNF chromatin remodeling complex,” Molecular and Cellular Biology , vol. 26, no. 7, pp. 2661–2674, 2006. View at Publisher · View at Google Scholar · View at Scopus S. Medjkane, E. Novikov, I. Versteege, and O. Delattre, “The tumor suppressor hSNF5/INI1 modulates cell growth and actin cytoskeleton organization,” Cancer Research , vol. 64, no. 10, pp. 3406–3413, 2004. View at Publisher · View at Google Scholar · View at Scopus I. Versteege, S. Medjkane, D. Rouillard, and O. Delattre, “A key role of the hSNF5/INI1 tumour suppressor in the control of the G1-S transition of the cell cycle,” Oncogene , vol. 21, no. 42, pp. 6403–6412, 2002. View at Publisher · View at Google Scholar · View at Scopus M. A. Swartz, J. Karth, D. T. Schneider, R. Rodriguez, J. B. Beckwith, and E. J. Perlman, “Renal medullary carcinoma: clinical, pathologic, immunohistochemical, and genetic analysis with pathogenetic implications,” Urology , vol. 60, no. 6, pp. 1083–1089, 2002. View at Publisher · View at Google Scholar · View at Scopus X. J. Yang, J. Sugimura, M. S. Tretiakova et al., “Gene expression profiling of renal medullary carcinoma: potential clinical relevance,” Cancer , vol. 100, no. 5, pp. 976–985, 2004. View at Publisher · View at Google Scholar · View at Scopus E. M. Schaeffer, T. J. Guzzo, K. A. Furge et al., “Renal medullary carcinoma: Molecular, pathological and clinical evidence for treatment with topoisomerase-inhibiting therapy,” BJU International , vol. 106, no. 1, pp. 62–65, 2010. View at Publisher · View at Google Scholar · View at Scopus var _gaq = _gaq || []; _gaq.push(['_setAccount', 'UA-8578054-2']); _gaq.push(['_trackPageview']); (function () { var ga = document.createElement('script'); ga.type = 'text/javascript'; ga.async = true; ga.src = ('https:' == document.location.protocol ? 'https://ssl' : 'http://www') + '.google-analytics.com/ga.js'; var s = document.getElementsByTagName('script')[0]; s.parentNode.insertBefore(ga, s); })();

Journal

Case Reports in Oncological MedicineHindawi Publishing Corporation

Published: Aug 19, 2014

There are no references for this article.