scholarly journals Safety of adjuvant gemcitabine plus cisplatin chemotherapy in a patient with bilateral ureteral cancer undergoing hemodialysis

Author(s):  
Yumiko Goto ◽  
Kent Kanao ◽  
Kazuhiro Matsumoto ◽  
Ikuo Kobayashi ◽  
Keishi Kajikawa ◽  
...  

AbstractAn 80 year old Japanese man with bilateral ureteral cancer underwent laparoscopic bilateral nephroureterectomy and lymph-node dissection. The pathological stage of the left and right ureteral tumors was pT3pN0M0. He received two courses of adjuvant gemcitabine and cisplatin chemotherapy while undergoing hemodialysis. The standard dose of gemcitabine and 50% of the standard dose of cisplatin were administered on the same day. Hemodialysis was started 6 h after gemcitabine administration and 1 h after cisplatin administration. The side effects were evaluated according to the Common Terminology Criteria for Adverse Events v4.0. In the first course, Grade 4 side effects including leukopenia, neutropenia, and thrombocytopenia were observed. He was treated with granulocyte colony-stimulating factor and platelet transfusion. Because the second course was administered without reducing the doses, granulocyte colony-stimulating factor was administered prophylactically, and Grade 4 side effects were reduced to Grade 3. Gemcitabine plus cisplatin chemotherapy can be administered safely in a patient with advanced ureteral cancer undergoing hemodialysis by adequately managing adverse events.

2020 ◽  
Author(s):  
Kitagawa Yusuke ◽  
Hiroki Osumi ◽  
Eiji Shinozaki ◽  
Yumiko Ota ◽  
Izuma Nakayama ◽  
...  

Abstract Background: This study aimed to evaluate the efficacy and safety of polyethylene glycol conjugated granulocyte colony-stimulating factor (PEG-G-CSF) for preventing neutropenia in metastatic colorectal cancer (mCRC) patients that received fluorouracil, leucovorin, oxaliplatin, and irinotecan (FOLFOXIRI) plus bevacizumab (Bev) in clinical practice. Methods: We retrospectively analyzed mCRC patients who received FOLFOXIRI plus Bev between December 2015 and December 2017. We evaluated the efficacy of PEG-G-CSF as preventing or treating grade 3/4 neutropenia, the overall response rate (ORR) according to the Response Evaluation Criteria in Solid Tumors version 1.1, progression-free survival (PFS), overall survival (OS), and adverse events of FOLFOXIRI plus Bev based on the Common Terminology Criteria for Adverse Events version 4.0. Results A total of 26 patients (median age 53.5 years) were included. The ORR rate was 65.3%, the median PFS was 9.6 months (7.2–16.9), and the median OS was 24.2 months (13.6–NA). Grade 3 or 4 neutropenia occurred in 53.8% of the patients, and febrile neutropenia occurred in 7.7%. PEG-G-CSF was given to 77.0% of the patients, including prophylactically (n = 9) and after the development of grade 3 or 4 neutropenia (n = 11). No patients experienced grade 3 or higher neutropenia after the administration of PEG-G-CSF. In seven of the nine patients who received PEG-G-CSF prophylactically (77.8%), no dose adjustment was required. Conclusions PEG-G-CSF is useful in preventing severe neutropenia in mCRC patients treated with FOLFOXIRI plus Bev.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5768-5768
Author(s):  
Bingzong Li ◽  
Ping Chen ◽  
Xueping Ge ◽  
Xiaohui Zhang ◽  
Jinxiang Fu

Abstract Background: Proteasome inhibition with bortezomib has revolutionized the treatment of multiple myeloma, but the vast majority of patients eventually develop clinical bortezomib resistance through poorly understood mechanisms. Long-lived plasma cells, which are supported by niche environments, are postulated to be an important mediator of resistance to chemotherapy and disease relapse. CXCR4/CXCL12 (SDF-1) axis plays an important role in the interaction of long-lived plasma cells and the microenvironment. Granulocyte-Colony Stimulating Factor (G-CSF) is reported to possess the potential to inhibit the expression of SDF-1, thus to suppress the CXCR4/SDF-1 axis and mobilize long-lived plasma cells from the niche environments. Bortezomib is reported to be able to eliminate both short- and long-lived plasma cells. In this trial we hypothesized that inhibition of the CXCR4/SDF-1 axis by G-CSF would disrupt the interaction of plasma cells with the environment and increase the sensitivity of myeloma cells to bortezomib. Methods: GBCD-001 (G-CSF, Bortezomib, Cyclophosphamide and Dexamethasone), a registered phase II study clinical trial (NCT02027220), engaged to both bortezomib-naïve and bortezomib-exposed myeloma patients. All the patients received G-BCD regimen at least one cycle. Dosing schedule consisted 28-day cycles of G-CSF 150µg, intracutaneously injection (IC) on days 0, 1, 7, 8, 14, 15, 21 and 22; Bortezomib 1.3mg/m2, intravenously injection (IV) on days 1, 8, 15 and 22; Cyclophosphamide 300 mg/m2, IV on days 1, 8, 15 and 22; and Dexamethasone 20mg/d, IV on days 1, 2, 8, 9, 15, 16, 22 and 23. Response was assessed by the IMWG criteria and toxicity was graded using the CTCAE v4.0. Results: From July 2013 to July 2014, 22 patients (10 females and 12 males; median age 63, range 44-80) meeting eligibility criteria have beed enrolled onto this study and are evaluated for response and toxicity, with 21 patients presenting with Durie-Salmon stage ¢ò/¢ó, 15 presenting with ISS stage ¢ò/¢ó disease. The median plasma cells is 32%, ranging from 5%-90%. Of the 22 pts, 19 are primary ones who had received no therapy and 3 are refractory ones who had received regimens including bortezomib. The response was listed in table 1. Of 19 primary pts, the ORR (¨RPR) was17/19 (89.5%) after one cycle and 10 pts received four cycles, among which, 7 (70%) achieved nCR/CR, 2 (20%) VGPR and 1 (10%) PR. Of 3 pts who received six cycles, all (100%) achieved CR. Of 3 refractory pts, the ORR was 2/3 (67%) after one cycle, 2 CR and 1 PR after 3 cycles. The most common adverse events observed included thrombocytopenia (40.9%, with 1 grade 4, 3 grade 3, 3 grade 2 and 2 grade 1), fatigue (40.9%, with 9 grade 1), neutropenia (22.7%, with 1 grade 4, 2 grade 2 and 2 grade 1), constipation (22.7%, with 2 grade 2 and 3 grade 1) and diarrhea (18.2%, with 1 grade 3, 1 grade 2 and 2 grade 1). Peripheral neuropathy possibly related to the drug was seen in 5 patients with grade 1 and 1 patient with grade 2, respectively. There were no deaths on study. Conclusions: In this study, among patients completing four cycles of G-BCD therapy, both primary and refractory ones, 100% have achieved PR or better. CR rate was 70% and 66.7% in primary pts and refractory ones, respectively. The adverse events were well tolerated and without special intervention. The study is currently recruiting more participants and updated results will be presented at the meeting. Large clinical studies and control studies are needed to replicate this promising results.Table 1 Response 1 cycle n/N (%) 4 cyclesn/N (%)6 cyclesn/N (%)PrimaryRefractoryPrimaryRefractoryPrimaryORR (¨RPR)17/19 (89.5)2/3(66.7)10/10 (100)3/3(100)3/3(100)nCR/CR1/19(5.3)07/10(70)2/3(66.7)3/3(100)VGPR1/19(5.3)02/10(20)00PR15/19(78.9)2/3(66.7)1/10(10)1/3(33.3)0 Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3984-3984
Author(s):  
Hua Wang ◽  
Yue Lu

Abstract Introduction: Although the considerable advances have been made in the treatment of acute myeloid leukemia (AML) in recent decades, only one third of patients (except for acute promyelocytic leukemia) can be cured , and most suffer relapse or primary refractory (R/R). However, there is no standard regimen for R/R AML patients after conventional chemotherapy.A regimen consisting of granulocyte colony-stimulating factor (G-CSF) priming the combination of low-dose cytarabine (Ara-C) and aclarubicin, termed the CAG regimen,was first reported by a Japanese group in 1995. It has been proved that cladribine is a attractive drug in AML because of their significant synergy with other chemotherapeutic agents and favorable toxicity profile. Efforts were made to improve the efficacy of the CAG regimen among institutions, including combining it with other chemotherapy drugs. Therefore, the purpose of this study is to prospectively evaluate efficacy and safety of C-CAG regimen (cladribine in combination with granulocyte colony-stimulating factor, low-dose cytarabine and aclarubicin) in patients with R/R AML(This study was registered at www.chictr.org, the Clinical Trial Registration Number was ChiCTR-OPC-16010166). Methods: Enrolment began in August 2016 for a Phase II single-center clinical trial . The patients in this arm will receive C-CAG regimen for salvage treatment,detailed as following:Cladribine 5mg/㎡,d1-5; G-CSF 300ug,d0-9; aclarubicin 10mg,d3-6;cytarabine 10mg/㎡ q12h, SC, d3-9;4 weeks a cycle.The patients are permitted to quit the study if complete remission(CR) is not achieved after 2 course of chemotherapy. If conditions were right,the patients achieving CR were recommended to receive allogeneic hematopoietic stem cell transplantation(HSCT). Otherwise, the patients were given for a total of six cycles unless there was disease progression or unacceptable side effects, or withdrawal of patient consent. Results: Until June 1, 2018, we have completed the enrolment of 15 patients with R/R AML. The main clinical characteristics of the 15 patients are presented in Table 1. The median age was 38 years and ranged from 21 to 72 years. The majority of patients presented with FAB subtype of M2 or M5. All patients(n=15) have begun treatment and are evaluable for response. Of the 15 patients, complete remission rates is 73.3% to C-CAG regimen. No patients received allogeneic transplantation due to scarcity of bone marrow donors or inability of the patients to afford the expensive medical treatment.At a median follow-up of 6 months (2-24), the median DFS and OS for all 15 patients were 8 and 12 months, respectively.The most common adverse effect was myelosuppression. The incidence of grade 3 or 4 hematological toxicity was 80.0%.The respective median duration of neutropenia (neutrophils < 0.5×109/L) was 12 days. The median time of platelet recovery over 50 × 109/L was 23 days.All patients presented an incidence of 13.3% for Grade 3 or 4 infectious toxicity. No treatment-related deaths occurred. Non-hematological side effects were mild. Grade 3 or 4 gastrointestinal side effects were rarely observed, owing to the prophylactic antiemetic drug administration. Conclusions: Preliminary data indicate that the C-CAG regimen chemotherapy is significantly effective against R/R AML with a high remission rate and a low hematological toxicity, thus serves as an alternative treatment for R/R AML. Table 1. Table 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4864-4864
Author(s):  
Antonio Lazzaro ◽  
Patrizia Bernuzzi ◽  
Elena Trabacchi ◽  
Daniele Vallisa ◽  
Annalisa Arcari ◽  
...  

Abstract Hematological toxicity during imatinib therapy in patients with chronic myelogenous leukaemia (CML) is common occurring in about 30–50% of cases. Grade 3 or 4 neutropenia occurs in 35–45% of patients with CML in chronic phase that receive imatinib. Often myelosuppression results in treatment interruption or dose reduction producing a negative effect on the efficacy of therapy. To reverse neutropenia induced by imatinib therapy in a patient with chronic-phase CML we used granulocyte-colony-stimulating factor (filgrastim) evaluating the response and the safety. A chronic-phase CML Philadelphia positive was diagnosed in a 42 years old woman presenting with elevated thrombocytosis (platelets 1082 x10E9/L), splenomegaly and mild leucocytosis (white cells 10.8 x10E9/L). Cytogenetic analysys showed a tipical bcl-abl rearrangement in 100% of mitoses. Hematological and clinical parameters were used to obtain a mild Sokal risk score (0.9). Imatinib was started at standard dose of 400 mg/day producing a partial cytogenetic response (bcl-abl in 30% of mitoses). After 8 weeks of therapy the patient showed a rapid decrease of platelets number to normal level but presented a grade 2 anemia (haemoglobin 9.6 gr/dl) and a grade 3 neutropenia (760 x10E9/L). The imatinib dose was first reduced to 300 mg/day without leucocytes number recovery. The therapy was then stopped and reintroduced at a dose of 200 mg/day after increase of total leucocytes number but the hematologic toxicity reappeared. A cytogenetic analysis performed after five months by the start of therapy showed a progression of leukemic clone (bcl-abl in 75% of mitoses). Granulocyte-colony-stimulating factor (filgrastim) was then introduced with a schedule of 300 microgr every seven days. The granulocyte-colony-stimulating factor therapy was well tolerated and the patient showed a disappearance of haematological toxicity after only 3 weeks. Neutrophils number was ever superior to 1.5 x10E9/L and haemoglobin level remained at a value of 11–11.5 gr/dl. The absence of hematologic toxicity permitted to increase the imatinib therapy at a standard dose of 400 mg/day. A cytogenetic analysis with fluorescent in situ hybridization (FISH) technique was performed after three months of standard dose (400 mg/day) and showed a complete cytogenetic response (bcl-abl rearrangement resulted absent in 100% of mitoses). Actually the patient is at the seventh months by the start of full dose of imatinib therapy and the neutrophils number is stable over 2.5 x10E9/L with a single dose of granulocyte-colony-stimulating factor every ten days. In conclusion, on the basis of the concept that the probability of response to imatinib is decreased for patients who developed myelosuppression and that needed a dose reduction of therapy, we believe that the use of granulocyte-colony-stimulating factor to reverse neutropenia represents a safe and efficacy method to contrast the hematologic toxicity and to permit an optimal imatinib terapeutic schedule.


Blood ◽  
1997 ◽  
Vol 90 (12) ◽  
pp. 4996-5001 ◽  
Author(s):  
Arnold Freedman ◽  
Donna Neuberg ◽  
Peter Mauch ◽  
John Gribben ◽  
Robert Soiffer ◽  
...  

Abstract Hematopoietic growth factors allow dose escalation of chemotherapy. This approach may potentially reduce the quality and quantity of hematopoietic stem cells. The capacity of stem cells recovered after dose intensification to support myeloablative therapy is unknown. In patients with previously untreated advanced follicular lymphoma, trilineage hematopoietic engraftment was compared in two sequential trials of induction therapy (standard dose cyclophosphamide, doxorubicin, vincristine, prednisone [CHOP] without growth factors or dose intensification CHOP supported by granulocyte colony-stimulating factor [G-CSF ]) followed by identical myeloablative therapy and autologous stem cell support. Neutrophil, platelet, and red blood cell (RBC) engraftment were compared on days 100, 180, and 360 after stem cell reinfusion. Despite similar patient characteristics including reinfusion of comparable numbers of marrow mononuclear cells, after stem cell transplantation, a highly significant prolongation of neutrophil and platelet engraftment was seen in patients who received high dose CHOP and G-CSF in comparison to standard dose CHOP. These findings suggest that dose intensified chemotherapy and G-CSF recruited stem cells into a proliferative phase and that G-CSF allowed retreatment at a time when stem cells were susceptible to damage by cytotoxic therapy. Such inadequate hematologic engraftment after myeloablative therapy might be avoided by either shortening the time that growth factor support is administered, lengthening the interval between cycles, or attempting to repetitively harvest additional stem cells either from the marrow or peripheral blood. Therefore, intensification of chemotherapy with growth factor support must be used with caution if stem cells are to be used to support myeloablative therapy.


1994 ◽  
Vol 12 (3) ◽  
pp. 483-488 ◽  
Author(s):  
P J Loehrer ◽  
P Elson ◽  
R Dreicer ◽  
R Hahn ◽  
C R Nichols ◽  
...  

PURPOSE This multicenter cooperative group phase I/II trial evaluated the toxicity and efficacy of escalated dosages of methotrexate, vinblastine, doxorubicin, and cisplatin (M-VAC) with recombinant human granulocyte colony-stimulating factor (rhG-CSF) in patients with advanced urothelial carcinoma. PATIENTS AND METHODS From November 1990 through October 1991, 35 patients with advanced urothelial cancer previously untreated with chemotherapy were treated with escalated dosages of M-VAC (M-VACII). In patients with prior pelvic radiotherapy, standard M-VAC (M-VACI) was administered plus rhG-CSF. For other patients, M-VACII dosages were methotrexate 40 mg/m2 (days 1, 15, and 22), vinblastine 4 mg/m2 (days 2, 15, and 22), doxorubicin 40 mg/m2 (day 2), and cisplatin 100 mg/m2 (day 2). In addition, rhG-CSF was administered at a dosage of 300 micrograms subcutaneously on days 4 to 11. Cycles were repeated every 4 weeks. For patients who tolerated the first course of therapy, subsequent escalation by 25% of all drugs was performed. RESULTS Six complete responses and 15 partial responses were observed (60%; 95% confidence interval, 42% to 76%). The median duration of response was 4.6 months, and the median survival time was 9.4 months (range, 0.5 to 23.5+). Twenty-eight of 35 patients experienced grade 3 or 4 leukopenia, including 14 patients who developed fever associated with neutropenia. Eight (23%) early deaths were observed. CONCLUSION This regimen (M-VACII) with escalated dosages of M-VAC was associated with significant toxicity and had no apparent benefit over M-VACI therapy with regard to complete response rate or survival. Further evaluation of the dose-intensity of the components of this regimen in this disease is likely to be of limited benefit to patients.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4111-4111
Author(s):  
Guangsheng He ◽  
Xiang Zhang ◽  
Wu Depei ◽  
Aining Sun ◽  
Zhengming Jin ◽  
...  

Abstract Abstract 4111 Objective Biphenotypic acute leukemia (BAL) involves both myeloid and lymphoid cells, and there is lack of uniformity in treatment at present. Optimal approach for therapy of BAL is unknown, and BAL usually becomes refractory to conventional chemotherapy. It was found that CAG regimen [low-dose cytosine arabinoside (Ara-C) plus aclarubicin with concurrent administration of granulocyte colony-stimulating factor (G-CSF)] is effective for both myeloid leukemia and refractory acute lymphoblastic leukemia. To explore the efficiency of CAG regimen for refractory BAL, 12 BAL patients who were failed to daunorubicin/mitoxantrone, cytosine arabinoside, vincristine, prednisone (D/MAOP) regimen, were treated by CAG. 8 were males and 4 was female, with ages ranging from 20 to 43 years (median=34 years). Immunophenotype of B lymphoid lineage/myeloid lineage showed in 8 patients, and T lymphoid lineage/myeloid lineage showed in 4 patients. The patients whose blast cells did not decrease 50% after induction regimen with lymphoid and/or myeloid drugs were defined as refractory cases. Methods 12 refractory BAL patients were treated by CAG regimen (cytosine arabinoside 10 mg/m2 subcutaneously every 12 hours, day 1-14; aclarubicin 5-7 mg/m2 intravenously daily, day 1-8; and concurrent use of granulocyte colony-stimulating factor 200μg·m-2·d-1 subcutaneously) from November 2002 to April 2009. The efficacy of the regimen was evaluated by response rate, and the side-effects was also measured. Results The major chemotherapy toxicity was bone marrow depression, mainly showing as pancytopenia. Median duration of absolute neutrophil count (ANC)<0.50×109/L and of platelet (PLT) count<20×109/L was 13 (range, 1-17) days and 1 day (range, 0-5 days), respectively. Median transfusion of red cells and PLT was 4 (range, 1.5-12) U and 20 (range, 0-40) U, respectively. Besides bone marrow depression, other side-effects such as infection, bleeding, nausea and vomiting also occurred during therapy. According to chemotherapy toxicity assessment of WHO, there were 7 patients confronting infection, among whom only 2 patient occurred III-‡W infection. No serious nausea vomiting or hepatic function damage happened. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 123 (23) ◽  
pp. 3655-3663 ◽  
Author(s):  
Michael A. Pulsipher ◽  
Pintip Chitphakdithai ◽  
Brent R. Logan ◽  
Willis H. Navarro ◽  
John E. Levine ◽  
...  

Key Points BM donors have a threefold higher risk for life-threatening, serious unexpected, or chronic adverse events vs PBSC donors (0.99% vs 0.31%). Donors receiving granulocyte colony-stimulating factor for PBSC collection had no evidence of increased risk for cancer, autoimmune illness, and stroke.


Stroke ◽  
2014 ◽  
Vol 45 (2) ◽  
pp. 623-626 ◽  
Author(s):  
Daniel-Christoph Wagner ◽  
Claudia Pösel ◽  
Isabell Schulz ◽  
Gerda Schicht ◽  
Johannes Boltze ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document