Neoadjuvant chemotherapy of docetaxel and carboplatin in patients with stage Ib2 to IIb nonsquamous cervix cancer of the uterus.

2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 5103-5103
Author(s):  
Shoji Nagao ◽  
Muneaki Shimada ◽  
Keiichi Fujiwara ◽  
Nobuhiro Takeshima ◽  
Ken Takizawa ◽  
...  

5103 Background: Non-squamous carcinoma of the uterine cervix has a resistance to radiation therapy and chemotherapy, and this is associated with worse prognosis compared with squamous carcinoma. This is a phase II study to assess an efficacy and safety of neoadjuvant chemotherapy of docetaxel and carboplatin (DC therapy) in patients with stage Ib2 to II non-squamous cervix cancer of the uterus. Methods: Patients with histologically confirmed, stage Ib2-II non-squamous uterine cervix cancer were received DC therapy prior to type III radical hysterectomy. IV doceaxel was administered at 60 mg/m2 followed by IV carboplatin administration based on AUC=6. The treatment was repeated every 21 days for a total of 1 to 3 cycles. Surgery was performed as soon as sufficient response was obtained. Results: Fifty-three women with non-squamous cervical carcinoma (FIGO stage Ib2,15; IIa, 7; IIb, 31) received DC therapy. Median age and tumor size were 49 years old (range: 27-71) and 52mm (range: 10-92), respectively. Fourty of 47 patients (74%) received 2 cycles of chemotherapy. Clinical response occurred in 69.8% of patients (complete response, 5; partial response, 32; stable disease, 15; disease progression, 1), and 96% (52/54) of patients completed the surgery. Incidences of grade 3/4 hematological toxicities were 98% (53/54) for neutrocytopenia, 4% (2/54) for thrombocytopenia, and 9% (5/54) for anemia. Febrile neutropenia was observed in three patients. Observed grade 3/4 non-hematological toxicities included were 5 for nausea, 2 for vomiting, 3 for constipation, 4 for allergic reaction. Median follow-up duration was 860 days with a range of 147- 1635 days. The 2-years progression-free survival rate and 2-years overall survival rate were 63% and 79% in stage IB2, 71% and 86% in stage IIA2, and 67% and 86% in stage IIB, respectively.Twenty patients recurred and twelve patients died. Conclusions: Neoadjuvant chemotherapy using DC therapy was well tolerated and had good response. It seems to be benefical in patients with satge Ib2 to II nonsquamous cervical cancer.

2010 ◽  
Vol 28 (5) ◽  
pp. 800-807 ◽  
Author(s):  
Antonio Palumbo ◽  
Francesca Gay ◽  
Patrizia Falco ◽  
Claudia Crippa ◽  
Vittorio Montefusco ◽  
...  

PurposeTo evaluate the effect of bortezomib as induction therapy before autologous transplantation, followed by lenalidomide as consolidation-maintenance in myeloma patients.Patients and MethodsNewly diagnosed patients age 65 to 75 years were eligible. Induction (bortezomib, doxorubicin, and dexamethasone [PAD]) included four 21-day cycles of bortezomib (1.3 mg/m2on days 1, 4, 8, and 11), pegylated liposomal doxorubicin (30 mg/m2on day 4), and dexamethasone (40 mg/d; cycle 1: days 1 to 4, 8 to 11, and 15 to 18; cycles 2 to 4: days 1 to 4). Autologous transplantation was tandem melphalan 100 mg/m2(MEL100) and stem-cell support. Consolidation included four 28-day cycles of lenalidomide (25 mg/d on days 1 to 21 every 28 days) plus prednisone (50 mg every other day), followed by maintenance with lenalidomide (LP-L; 10 mg/d on days 1 to 21) until relapse. Primary end points were safety (incidence of grade 3 to 4 adverse events [AEs]) and efficacy (response rate).ResultsA total of 102 patients were enrolled. In a per-protocol analysis, after PAD, 58% of patients had very good partial response (VGPR) or better, including 13% with complete response (CR); after MEL100, 82% of patients had at least VGPR and 38% had CR; and after LP-L, 86% of patients had at least VGPR and 66% had CR. After median follow-up time of 21 months, the 2-year progression-free survival rate was 69%, and the 2-year overall survival rate was 86%. During induction, treatment-related mortality was 3%; grade 3 to 4 AEs included thrombocytopenia (17%), neutropenia (10%), peripheral neuropathy (16%), and pneumonia (10%). During consolidation-maintenance, grade 3 to 4 AEs were neutropenia (16%), thrombocytopenia (6%), pneumonia (5%), and cutaneous rash (4%).ConclusionBortezomib as induction before autologous transplantation, followed by lenalidomide as consolidation-maintenance, is an effective regimen.


2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 4027-4027 ◽  
Author(s):  
C. Fuchs ◽  
J. Marshall ◽  
E. Mitchell ◽  
R. Wierzbicki ◽  
V. Ganju ◽  
...  

4027 Background: This multicenter, randomized study assessed efficacy & safety for irinotecan/fluoropyrimidines combinations in previously untreated mCRC. Methods: Pts were randomized to: infusional FOLFIRI, modified bolus IFL (mIFL), or CapeIri; and concurrent celecoxib or placebo in a double-blind fashion. The protocol was amended in April 2004: bevacizumab (bev) was added to the FOLFIRI and mIFL arms, whereas CapeIri was discontinued. Period 1 (P1) and Period 2 (P2) designate subjects enrolled before or after the amendment. Initial efficacy & safety analyses were reported at ASCO ’06. We now report follow-up of 46 months for P1 and 31 months for P2. Results: 430 pts were treated in P1 and 117 pts in P2. Baseline characteristics and post-study treatment were balanced. P1 results: Median progression free survival (PFS) was 7.6 mos for FOLFIRI; 5.9 mos for mIFL (p=0.004); and 5.8 mos for CapeIri (p=0.015). Median overall survival (OS) was 23.1 mos for FOLFIRI; 17.6 mos for mIFL (p=0.087); and 18.9 mos for CapeIri (p=0.27). One-year survival rate favored FOLFIRI (75%) compared to either mIFL (65%) or CapeIri (66%). Overall Response Rate (ORR) was 47% in FOLFIRI, 43% in mIFL, 39% in CapeIri (not significantly different). P2 results: Median PFS was 11.2 mos for FOLFIRI+bev and 8.3 mos for mIFL+bev (p=0.28). Median OS was not reached for FOLFIRI+bev but was 19.2 mos for mIFL+bev (p=0.007). One-year survival rate favored FOLFIRI+bev (87%) when compared to mIFL+bev (61%). ORR was 58% for FOLFIRI+bev and 54% for mIFL+bev (p=0.73). Common grade = 3 AEs are listed below. Celecoxib did not impact safety or efficacy. Conclusions: First line FOLFIRI or FOLFIRI+bev were superior to their comparators and show favorable results in survival and tolerability in untreated mCRC. Median survival for FOLFIRI+bev has not been reached. [Table: see text] No significant financial relationships to disclose.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. 4040-4040 ◽  
Author(s):  
Heinz-Josef Lenz ◽  
Sara Lonardi ◽  
Vittorina Zagonel ◽  
Eric Van Cutsem ◽  
M. Luisa Limon ◽  
...  

4040 Background: In the phase 2 CheckMate 142 trial, NIVO + low-dose IPI had robust, durable clinical benefit and was well tolerated as 1L therapy for MSI-H/dMMR mCRC (median follow-up 13.8 months [mo; range, 9–19]; Lenz et al. Ann Oncol 2018;29:LBA18). Longer follow-up is presented here. Methods: Patients (pts) with MSI-H/dMMR mCRC and no prior treatment for metastatic disease received NIVO 3 mg/kg Q2W + low-dose IPI 1 mg/kg Q6W until disease progression or discontinuation. The primary endpoint was investigator-assessed (INV) objective response rate (ORR) per RECIST v1.1. Results: In 45 pts with median follow-up of 29.0 mo, ORR (95% CI) increased to 69% (53–82) (Table) from 60% (44.3–74.3); complete response (CR) rate increased to 13% from 7%. The concordance rate of INV and blinded independent central review was 89%. Median duration of response (DOR) was not reached (Table). Median progression-free survival (PFS) and overall survival (OS) were not reached, and 24-mo rates were 74% and 79%, respectively (Table). Nineteen pts discontinued study treatment without subsequent therapy. An analysis of tumor response post discontinuation will be presented. Ten (22%) pts had grade 3–4 treatment-related adverse events (TRAEs); 3 (7%) had grade 3–4 TRAEs leading to discontinuation. Conclusions: NIVO + low-dose IPI continued to show robust, durable clinical benefit with a deepening of response, and was well tolerated with no new safety signals identified with longer follow-up. NIVO + low-dose IPI may represent a new 1L therapy option for pts with MSI-H/dMMR mCRC. Clinical trial information: NTC02060188 . [Table: see text]


2014 ◽  
Vol 32 (7) ◽  
pp. 634-640 ◽  
Author(s):  
Antonio Palumbo ◽  
Sara Bringhen ◽  
Alessandra Larocca ◽  
Davide Rossi ◽  
Francesco Di Raimondo ◽  
...  

Purpose Bortezomib-melphalan-prednisone (VMP) has improved overall survival in multiple myeloma. This randomized trial compared VMP plus thalidomide (VMPT) induction followed by bortezomib-thalidomide maintenance (VMPT-VT) with VMP in patients with newly diagnosed multiple myeloma. Patients and Methods We randomly assigned 511 patients who were not eligible for transplantation to receive VMPT-VT (nine 5-week cycles of VMPT followed by 2 years of VT maintenance) or VMP (nine 5-week cycles without maintenance). Results In the initial analysis with a median follow-up of 23 months, VMPT-VT improved complete response rate from 24% to 38% and 3-year progression-free-survival (PFS) from 41% to 56% compared with VMP. In this analysis, median follow-up was 54 months. The median PFS was significantly longer with VMPT-VT (35.3 months) than with VMP (24.8 months; hazard ratio [HR], 0.58; P < .001). The time to next therapy was 46.6 months in the VMPT-VT group and 27.8 months in the VMP group (HR, 0.52; P < .001). The 5-year overall survival (OS) was greater with VMPT-VT (61%) than with VMP (51%; HR, 0.70; P = .01). Survival from relapse was identical in both groups (HR, 0.92; P = .63). In the VMPT-VT group, the most frequent grade 3 to 4 adverse events included neutropenia (38%), thrombocytopenia (22%), peripheral neuropathy (11%), and cardiologic events (11%). All of these, except for thrombocytopenia, were significantly more frequent in the VMPT-VT patients. Conclusion Bortezomib and thalidomide significantly improved OS in multiple myeloma patients not eligible for transplantation.


Author(s):  
Fransisca Noela ◽  
Kartiwa H Nuryanto

Objective: To describe the incidence of ovarian cancer and its characteristic in Dr. Cipto Mangunkusumo Hospital in the last 5 years. Method: This was cross sectional study design. The data was collected from Gynecology Oncology Division Cancer Registry and Dr. Cipto Mangunkusumo Hospital medical record from January 2009 to December 2013; follow up was performed to know the 4-years survival rate. Result: There were 98 subjects in this study. The majority incidence of ovarian cancer was 45-54 years old (33.6%); the incidence of ovarian cancer decreased with the increased number of parity; the majority histotype was epithelial (76.5%); and most of them were diagnosed on advanced stage (55.1%). The 4-year survival rate for epithelial type was 77%; germinal type was 83.3%; and stromal type was 100%. Based on therapy, the 4-year survival rate was 84.1% for surgical only; 83.3% in adjuvant chemotherapy group; and 68.4% in neoadjuvant chemotherapy. In the group of adjuvant chemotherapy, there was 63% patients with complete response and 41.2% patients with complete response in neoadjuvant chemotherapy. Conclusion: The highest incidence of ovarian cancer in Dr. Cipto Mangunkusumo Hospital belongs to the age of reproductive women (≤ 55 years old) with the highest incidence occurs in nulliparity women. Most of the ovarian cancer cases are diagnosed in advanced stage (stage III-IV). [Indones J Obstet Gynecol 2016; 4-2: 101-106] Keywords: age, histotype, ovarian cancer, parity, response, stage, survival, treatment


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. 2037-2037
Author(s):  
Giuseppe Lamberti ◽  
Enrico Franceschi ◽  
Alicia Tosoni ◽  
Santino Minichillo ◽  
Monica Di Battista ◽  
...  

2037 Background: Medulloblastoma is extremely rare in adults and, therefore, it is difficult to accrual patients in clinical trials. Radical surgery and radiotherapy (RT) provide a significant control of disease. Nevertheless, about 25% of average-risk patients have a relapse and die because of disease progression. The role of chemotherapy (CT) after standard RT for average-risk adult patients remains controversial. Methods: We analyzed 48 average-risk patients according to Chang classification diagnosed from 1988 to 2016. Median age was 29 years (range 16-61), M/F ratio was 26 (54.2%)/22 (45.8%). Fifteen patients had classic medulloblastoma (31.3%), 15 patients had desmoplastic medulloblastoma (31.3%), 5 patients had extensive nodularity (10.4%) and 2 patients had large cells/anaplastic histology (4.2%). The patients were homogeneously distributed in the two groups: 24 (50%) received adjuvant RT alone and 24 (50%) received RT + CT that consisted in a platinum-etoposide based combination. Results: After a median follow-up of 12.5 years, CT increases progression-free survival rate at 15 years (PFS-15 82.3 ± 8.0% in RT-CT group vs. 38.5% ± 13.0% in RT group p = 0.05) and overall survival rate at 15 years (OS-15 89.3% 7.2% vs. 52.0% 13.1%, p = 0.02). Among patients receiving CT, the reported grade ≥ 3 adverse events were: 9 cases of neutropenia; 6 cases of G3 neutropenia (25%) and 3 cases of G4 neutropenia (13%), 1 case of G3 thrombocytopenia (4%) and 2 cases of G3 nausea (8%). Conclusions: Our study with a long follow up period suggests that adding adjuvant chemotherapy to RT might improve PFS and OS in average-risk adult medulloblastoma patients.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 210-210
Author(s):  
Thomas S Lin ◽  
Kathleen A Donohue ◽  
John C. Byrd ◽  
Margaret S Lucas ◽  
Eva Hoke ◽  
...  

Abstract Abstract 210 Background: Alemtuzumab (Campath-1H) is approved for the treatment of CLL. CALGB sought to determine whether alemtuzumab can improve the CR rate and eradicate MRD after induction chemoimmunotherapy by performing a phase II study administering FR followed by alemtuzumab consolidation to previously untreated, symptomatic CLL patients (pts). We previously reported preliminary toxicity data (Lin et al. ASH 2007) and now report final toxicity and response data. Methods: Pts received fludarabine 25 mg/m2 IV on days 1–5, and rituximab 50 mg/m2 IV on day 1, 325 mg/m2 on day 3, and 375 mg/m2 on day 5 of cycle 1 and then only on day 1 of cycles 2–6, repeated every 28 days for up to 6 cycles. Four months after the last fludarabine dose, pts with stable (SD) or responsive disease by NCI 96 criteria received SC alemtuzumab 3 mg on day 1, 10 mg on day 3, and 30 mg on day 5, and then thrice weekly thereafter for 6 weeks (18 total doses). Pts received standard Pneumocystis (PCP) and Varicella zoster virus prophylaxis and were monitored weekly by PCR for Cytomegalovirus (CMV) viremia. When unacceptable serious infectious toxicity was noted in pts who received alemtuzumab after achieving CR from FR induction, the study was amended so that only PR or SD pts received alemtuzumab after FR. Results: Median age of pts (n=102) was 61 years (range, 23–82), 74% were male, and 30% were Rai stage III/IV. FR was well tolerated; 93% of pts received at least 3 cycles, and 77% completed all 6 cycles. Overall, complete and partial response (OR, CR, PR) rates after FR induction were 90%, 29% and 61%, and 15% were MRD negative by flow cytometry. Fifty-eight pts received alemtuzumab, and 42 (72%) completed the planned 6 weeks of therapy. OR, CR, and PR rates after alemtuzumab (n=58) were 91%, 66% and 26%, and 50% were MRD negative. Twenty-eight of 45 pts (62%) in PR after FR who received alemtuzumab attained CR. Of 11 pts in CR after FR who received alemtuzumab, 5 were MRD negative prior to consolidation and 3 of the other 6 converted to MRD negative afterwards. By intent-to-treat for all patients enrolled, OR, CR, and MRD negativity were attained by 90%, 57% and 42% of pts. With a median follow up of 34 months, median progression free survival (PFS) was 37 months (95% CI, 33–43 months); PFS was 73% and overall survival (OS) 86% at 2 years. Two-year PFS (76% vs 70%, p=0.54) and OS (84% vs 88%, p=0.89) were similar for pts who did and did not receive alemtuzumab. Similarly, there were no differences in PFS or OS among the 30 pts in CR after FR whether or not they received alemtuzumab, although the numbers were small. Grade 3–4 neutropenia and thrombocytopenia were observed in 43% and 19% of pts during alemtuzumab therapy. Grade 3–4 non-hematologic toxicity was observed in 41% of pts, including 19% infections and 19% febrile neutropenia, during alemtuzumab therapy. As we previously reported, 5 pts in CR after FR who received alemtuzumab died from infections (viral meningitis, Listeria meningitis, Legionella pneumonia, CMV and PCP pneumonia), and one pt in PR after FR who received alemtuzumab died of Epstein-Barr (EBV) viremia without evidence of EBV lymphoma. These grade 5 toxicities occurred both during and for up to 7 months after alemtuzumab therapy. Conclusions: Alemtuzumab consolidation improved the CR and MRD negative rates after FR induction. However, alemtuzumab consolidation resulted in significant toxicity, particularly severe infections in pts who achieved a CR after FR induction. Longer follow up is needed to determine if the improved CR and MRD negative rates following alemtuzumab consolidation will eventually result in improved PFS or OS, especially among pts who achieved a PR after FR induction. Disclosures: Lin: GlaxoSmithkline: Consultancy, Employment; Genentech: Consultancy; Bayer: Consultancy. Off Label Use: Use of alemtuzumab as consolidation therapy. Byrd:Genentech: Consultancy, Research Funding. Link:Genentech: Consultancy. Rai:Genentech: Consultancy; Bayer: Consultancy.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. e17013-e17013
Author(s):  
Nabeel Naban ◽  
Gesa Tiemeier ◽  
Arwa Kocache ◽  
Rory Taylor ◽  
Georgina Keogh ◽  
...  

e17013 Background: The Covid19 pandemic has imposed risks to healthcare delivery for all cancer pts including young adults diagnosed with a GCT. Here, we examine Emergency EP (EmEP, Cohort A) and a novel 1-day Escalated-dose EP (EscEP, Cohort B) administered every 2 weeks as upfront or first-relapse therapy to minimise hospital days. Methods: Single-centre analysis in GCT pts receiving emergency and routine chemotherapy following our first National lockdown on 23rd March 2020. In Cohort A, eligible pts receive EmEP E100mg/m2, P20mg/m2 prior to EscEP within the acute setting as previously defined. In Cohort B, EscEP E500mg/m2, P60mg/m2 is compared to standard care chemotherapy BEP or POMB-ACE. Data collection includes demographics, treatment details, clinical outcome and Covid19 complications. Results: To date, we have accrued 19 pts with a median age 32 (range 18 to 65). In Cohort A, 7 pts with symptomatic high-volume disease received a median 1 cycle EmEP, 6/7 for a new GCT diagnosis and 1/7 for a first relapse, including 2 males with IGCCCG intermediate-prognosis disease, 5 females with FIGO Stage III (n = 2) and IV (n = 3) disease. One pt required higher-level support for organ dysfunction at presentation. In Cohort B, 19 pts including 7 pts from Cohort A received a median 4 cycles EscEP: 8 males (2 seminomas, 6 non-seminomas; IGCCCG good-prognosis in 4, intermediate-prognosis in 2, poor-prognosis in 2), 11 females (1 dysgerminoma, 10 non-dysgerminomas; FIGO Stage Ic in 6, III in 2, IV in 3). A majority (14/19, 74%) received EscEP for a new cancer diagnosis, 5/19 (26%) for a first relapse. Total median hospital stay: EscEP 5 days, BEP 21 days, POMB-ACE 28 days. Grade 3/4 neutropenic events: EscEP 28%, BEP 50%, POMB-ACE 43%. From April 2020, 16/16 pts were SARS-CoV-2 screened prior to each cycle: 3/19 (16%) testing positive, one prior to intubation and ventilation, 2 with asymptomatic infection. For Cohort B, at median follow-up 121 days (range 9-323 days), we have observed a complete response in 10 pts (53%), partial response in 3 pts (16%) and disease progression in 1 pt (5%). Five pts are still on treatment (26%). All pts remain alive. Conclusions: EmEP and EscEP represent safe options during the pandemic that minimise myelosuppression and total length in hospital days whilst bypassing the potential pulmonary toxicity from Bleomycin. Further follow-up will inform on long-term efficacy including a multicentre evaluation.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 13578-13578
Author(s):  
C. Gennatas ◽  
V. Michalaki ◽  
S. Gennatas ◽  
A. Kondi-Paphiti ◽  
D. Voros ◽  
...  

13578 Background: Capecitabine is an oral fluoropyrimidine with superior activity and safety compared with bolus 5-FU/LV in metastatic colorectal cancer (CRC). The aim of this study was to evaluate the efficacy and safety of a combination of capecitabine and oxaliplatin as first-line chemotherapy in patients with advanced CRC. Methods: Fourty-six patients (26 men and 20 women) with metastatic CRC entered this study. All patients were treated with capecitabine (1,000mg/m2 p.o.twice daily, days 1–14) and oxaliplatin (130mg/m2 on day 1). Cycles were repeated every 21 days until disease progression or unacceptable toxicity. Baseline characteristics: Median age 61 years (range 32–74), main sites of metastasis: Liver 32 patients (70%), liver and lungs 4 patients (9%), lungs 3 patients (6%), other sites 7 patients (15%). Results: 2 patients (4%) achieved complete response (CR), 17 patients (37%) achieved partial response (PR) and 7 patients (15%) attained stable disease (SD). With a median follow-up of 22 months the progression free survival was 7.5 months and overall survival was 19.0 months. All patients were assessable for toxicities. The most commonly encountered adverse events were from the gastrointestinal system (all grades 48%, grade 3, 6%). Neither toxic death nor life-threatening febrile neutropenia were reported. Conclusions: The combination of capecitabine and oxaliplatin is a convenient regimen in patients with advanced CRC that is associated with considerable efficacy and limited toxicity. No significant financial relationships to disclose.


2012 ◽  
Vol 30 (5_suppl) ◽  
pp. 307-307
Author(s):  
Sarah P. Psutka ◽  
Aria F. Olumi ◽  
Adam S. Feldman ◽  
Philip James Saylor ◽  
Donald S. Kaufman ◽  
...  

307 Background: Neoadjuvant chemotherapy (NC) with MVAC (methotrexate, vinblastine, doxorubicin, and cisplatin) improves survival in muscle invasive urothelial cancer (MI−UC) with patients who achieve pathologic complete response (PCR) following radical cystectomy (RC). Gemcitabine/cisplatin (GC) NC is increasingly employed due to lower toxicity, however, its effectiveness as neoadjuvant therapy is controversial and multiple studies have reported poor utilization of this therapy, despite recent recommendations advocating for broader use. We describe pathologic and clinical outcomes following NC and RC. Methods: We retrospectively evaluated patients with MI−UC who received NC between 2003 and 2011 (n=38). Those who were treated with neoadjuvant radiation therapy (n= 15) were excluded. We compared initial clinical stage at surgery to final pathological stage and assessed overall−median progression free−survival. Mean follow−up was 25 months (SD 21.6, range 3 –76 months). Results: Twenty-three patients who received NC were included. Nineteen patients (82.6%) were treated with GC, 3 (13.0%) with MVAC, and 1 (4.3%) with gemcitabine/paclitaxel. The median time from start of NC to RC was 119 days (IQR 98.5−146). 10/19 (52.6%) patients treated with GC achieved PCR (pT0) from clinical stage T2 (n=5), cT3 (n=2) and cT4 (n=3), and 6 (31.2%) were downstaged to pT1 and pTIS from cT2. Two patients treated with MVAC were downstaged to pT1 and one achieved PCR. Median recurrence-free survival was 13 months (IQR 6−19 months) with 8 patients developing recurrent or metastatic UC following RC. At median follow-up of 19 months (IQR 8−31.2 months, Range 1−71 months), 15/23 (65.2%) patients were disease free, all of whom had received GC NC prior to cystectomy. Conclusions: Neoadjuvant GC for MI−UC was associated with a 52.6% PCR rate at RC and was well tolerated. These data compare favorably with published data on GC and MVAC as NC, and warrant further study.


Sign in / Sign up

Export Citation Format

Share Document