Carboplatin chemical structure
Find information on thousands of medical conditions and prescription drugs.

Carboplatin

Carboplatin is a chemotherapy drug used against some form of cancer. It was introduced in the late 1980s and has since gained popularity in clinical treatment due to its vastly reduced side-effects compared to its parent compound cisplatin. Cisplatin and carboplatin, as well as oxaliplatin, are classified as DNA alkylating agents. more...

Home
Diseases
Medicines
A
B
C
Cabergoline
Caduet
Cafergot
Caffeine
Calan
Calciparine
Calcitonin
Calcitriol
Calcium folinate
Campath
Camptosar
Camptosar
Cancidas
Candesartan
Cannabinol
Capecitabine
Capoten
Captohexal
Captopril
Carbachol
Carbadox
Carbamazepine
Carbatrol
Carbenicillin
Carbidopa
Carbimazole
Carboplatin
Cardinorm
Cardiolite
Cardizem
Cardura
Carfentanil
Carisoprodol
Carnitine
Carvedilol
Casodex
Cataflam
Catapres
Cathine
Cathinone
Caverject
Ceclor
Cefacetrile
Cefaclor
Cefaclor
Cefadroxil
Cefazolin
Cefepime
Cefixime
Cefotan
Cefotaxime
Cefotetan
Cefpodoxime
Cefprozil
Ceftazidime
Ceftriaxone
Ceftriaxone
Cefuroxime
Cefuroxime
Cefzil
Celebrex
Celexa
Cellcept
Cephalexin
Cerebyx
Cerivastatin
Cerumenex
Cetirizine
Cetrimide
Chenodeoxycholic acid
Chloralose
Chlorambucil
Chloramphenicol
Chlordiazepoxide
Chlorhexidine
Chloropyramine
Chloroquine
Chloroxylenol
Chlorphenamine
Chlorpromazine
Chlorpropamide
Chlorprothixene
Chlortalidone
Chlortetracycline
Cholac
Cholybar
Choriogonadotropin alfa
Chorionic gonadotropin
Chymotrypsin
Cialis
Ciclopirox
Cicloral
Ciclosporin
Cidofovir
Ciglitazone
Cilastatin
Cilostazol
Cimehexal
Cimetidine
Cinchophen
Cinnarizine
Cipro
Ciprofloxacin
Cisapride
Cisplatin
Citalopram
Citicoline
Cladribine
Clamoxyquine
Clarinex
Clarithromycin
Claritin
Clavulanic acid
Clemastine
Clenbuterol
Climara
Clindamycin
Clioquinol
Clobazam
Clobetasol
Clofazimine
Clomhexal
Clomid
Clomifene
Clomipramine
Clonazepam
Clonidine
Clopidogrel
Clotrimazole
Cloxacillin
Clozapine
Clozaril
Cocarboxylase
Cogentin
Colistin
Colyte
Combivent
Commit
Compazine
Concerta
Copaxone
Cordarone
Coreg
Corgard
Corticotropin
Cortisone
Cotinine
Cotrim
Coumadin
Cozaar
Crestor
Crospovidone
Cuprimine
Cyanocobalamin
Cyclessa
Cyclizine
Cyclobenzaprine
Cyclopentolate
Cyclophosphamide
Cyclopropane
Cylert
Cyproterone
Cystagon
Cysteine
Cytarabine
Cytotec
Cytovene
Isotretinoin
D
E
F
G
H
I
J
K
L
M
N
O
P
Q
R
S
T
U
V
W
X
Y
Z

History

Bristol-Myers Squibb gained FDA approval for carboplatin, under the brand name Paraplatin, in March 1989. The drug went generic in October 2004.

Suppliers

Bristol-Myers Squibb continues to market Paraplatin. There are also generic versions of the drug available from APP, Bedford, Sicor, Mayne Pharma, Pharmachemie, Pliva, Sandoz, Spectrum.

Pharmacology

Chemistry

Carboplatin differs from cisplatin in that it has a closed cyclobutane dicarboxylate (CBDCA) moiety on its leaving arm in contrast to the readily leaving chloro groups. This results in very different DNA binding kinetics, though it forms the same reaction products in vitro at equivalent doses with cisplatin. However, recent studies provide a new caveat on the DNA binding molecular mechanisms with the possibility of being activated by nucleophiles (as opposed to cisplatin), before forming the toxic adducts. There are also results to show that cisplatin and carboplatin cause different morphological changes in MCF-7 cell lines while exerting their cytotoxic behaviour.

Mode of action

Two theories exist to explain the molecular mechanism of action of carboplatin with DNA.

  • Aquation, or the like-cisplatin hypothesis.
  • Activation, or the unlike-cisplatin hypothesis.

The former is more accepted owing to the similarity of the leaving groups with its predecessor cisplatin, while the latter hypothesis envisages a biologically activation mechanism to release the active Pt2+ species.

Side-effects

The largest benefit of using carboplatin over cisplatin is the reduction of side effects; particularly the elimination of cisplatin's nephrotoxic effects. This is due in part to the added stability of carboplatin in the bloodstream, which prevents proteins from binding to it. This in turn reduces the amount of these protein-carboplatin complexes to be excreted. The lower excretion rate of carboplatin means that more is retained in the body, and hence its effects are longer lasting (a retention half-life of 30 hours for carboplatin, compared to 1.5-3.6 hours in the case of cisplatin).

There are no known ototoxic effects from carboplatin. Nausea and vomiting are less severe and more easily controlled, compared to the incessant vomiting and antiperistalsis that some patients using cisplatin may experience. Carboplatin has also proven effective in some strains of cancer that may not be susceptible to cisplatin, including germ-line cell, small and non-small cell lung, ovary, and bladder cancers, as well as acute leukemia.

The main drawback of carboplatin is its myelosuppressive effects. This causes the blood cell and platelet output of bone marrow in the body to decrease quite dramatically, sometimes as low as 10% of its usual production levels. The nadir of this myelosuppression usually occurs 21-28 days after the first treatment, after which the blood cell and platelet levels in the blood begin to stabilize, often coming close to its pre-carboplatin levels. This decrease in white blood cells (neutropenia) causes many complications, most notably infection by opportunistic organisms. This necessitates readmission to hospital and treatment with antibiotics.

Read more at Wikipedia.org


[List your site here Free!]


Double-cycle, high-dose ifosfamide, carboplatin, and etoposide followed by peripheral blood stem-cell transplantation for small cell lung cancer
From CHEST, 10/1/05 by Yoshinobu Iwasaki

Purpose: To determine the tolerability and feasibility of double-cycle, high-dose chemotherapy followed by peripheral blood stem-cell transplantation (PBSCT) after conventional chemotherapy or chemoradiotherapy for small cell lung cancer (SCLC).

Patients and methods: Patients with previously untreated SCLC received two cycles of cisplatin, 80 mg/[m.sup.2], and etoposide, 300 mg/[m.sup.2] (cisplatin-etoposide [PE]). Later, they were administered high-dose etoposide, 1,500 mg/[m.sup.2], followed by granulocyte colony-stimulating factor for collection of peripheral blood stem cells. After two additional cycles of PE, the patients received high-dose ifosfamide, 10 g/[m.sup.2], carboplatin, 1,200 mg/[m.sup.2], and etoposide, 1,000 mg/[m.sup.2] (ifosfamide-carboplatin-etoposide [ICE]) followed by PBSCT twice at 3-month to 4-month intervals. Patients with limited disease (LD) concurrently received 50 Gy of irradiation with the last two cycles of PE.

Results: Eighteen patients, including 11 patients with LD, were enrolled. Fifteen patients could receive high-dose ICE followed by PBSCT twice, and 3 patients could receive it once. The median number of CD34+ cells collected was 13.11 x [10.sup.6]/kg. The median numbers of days to neutrophil counts [greater than or equal to] 500/[micro]L and platelet counts [greater than or equal to]50,000/[micro]L were 10 days and 14.5 days after the first PBSCT, and 10 days and 15 days after the second PBSCT, respectively. Grade 3 diarrhea occurred in one cycle, and grade 3 renal toxicity occurred in two cycles. The overall response rate was 100%, with an 83.3% rate of complete or near-complete response. The 2-year and 5-year survival rates were 72% and 55% in patients with LD and 43% and 0% in patients with extensive disease, respectively. Conclusion: Double-cycle, high-dose ICE therapy followed by PBSCT is tolerable and feasible even after conventional chemotherapy or chemoradiotherapy in patients with SCLC.

Key words: high-dose chemotherapy; peripheral blood stem-cell transplantation; small cell lung cancer

Abbreviations: ABMT = autologous bone marrow transplantation; CR = complete response; ED = extensive disease; G-CSF = granulocyte colony-stimulating factor; ICE = ifosfamide-carboplatin-etoposide; LD = limited disease; nCR = near complete response; PBSC = peripheral blood stem cell; PBSCT = peripheral blood stem-cell transplantation; PE = cisplatin-etoposide; PR = partial response; SCLC = small cell lung cancer

**********

Combination chemotherapy with cisplatin and etoposide (cisplatin-etoposide [PE]) has long been the mainstay of treatment for small cell lung cancer (SCLC). Response rates in extensive disease (ED) range from 51 to 78%, including a 7 to 13% rate of complete response (CR) and a median survival of 8 to 9 months. In patients with limited disease (LD), the CR rate is 16 to 18%, with a median survival of 11.7 to 12.4 months. (1-3)

Given the exquisite initial sensitivity of SCLC to chemotherapy and the high rate of relapse, some studies have attempted to improve survival by increasing dose intensity. Within conventional ranges, dose intensity can be increased with the support of hematopoietic growth factors. One approach to treatment is the rapid sequencing of several active agents over a short period. (4-6) Another approach is the use of higher doses of chemotherapy, particularly during the first four cycles of treatment. (7,8) The dose intensity of chemotherapy has also been increased by reducing the interval between chemotherapy cycles with the use of hematopoietic growth factors. (9-12) However, whether such approaches improve survival as compared with standard therapy remains controversial.

Autologous bone marrow transplantation (ABMT) or peripheral blood stem cell transplantation (PBSCT) have been used in many studies to control the hematologic toxicity of high-dose chemotherapy, thereby permitting a further increase in dose intensity. Most studies have evaluated late intensification strategies, in which a single course of intensive chemotherapy is administered to consolidate the response to standard treatment. A combined analysis (13) revealed no apparent improvement in survival, even though the percentage of complete responders almost doubled. Another study (14) proposed that late-intensification chemotherapy might improve long-term survival in a significant proportion of complete responders with LD. To date, only one randomized, phase III trial (15) of high-dose chemotherapy followed by ABMT or PBSCT has been completed in patients with SCLC. Significant differences favoring high-dose chemotherapy were seen with respect to relapse-free survival, (15) suggesting that double-cycle, high-dose chemotherapy with ABMT or PBSCT might prolong survival by reducing the relapse rate. Multiple cycles of high-dose chemotherapy have been used as first-line therapy, (16) not for late intensification. Recently, the safety of high-dose chemotherapy has been enhanced by improvements in supportive care, including the use of hematopoietic growth factors and peripheral blood stem cells (PBSCs).

The most recent studies have evaluated ifosfamide, carboplatin, and etoposide (ifosfamide-carboplatin-etoposide [ICE]) therapy, considered to have a favorable therapeutic index. (16-19) A steep dose response coupled with synergistic antitumor activity and a favorable spectrum of nonhematopoietic toxicity makes this combination a natural candidate for high-dose therapy. (20) A phase I dose-escalation study of high-dose ICE therapy followed by ABMT or PBSCT was conducted by Fields et al (21) to determine the maximum tolerated dose. They reported that the maximum tolerated dose of ICE was 20,100 mg/[m.sup.2] of ifosfamide, 1,800 mg/[m.sup.2] of carboplatin, and 3,000 mg/[m.sup.2] of etoposide. The dose-limiting toxicities of ICE were CNS toxicity and acute renal failure. Leyvraz et al (16) showed that three cycles of high-dose ICE therapy could be safely administered as first-line therapy. In their study, three cycles of high-dose ICE therapy with 10 g/[m.sup.2] of ifosfamide, 1,200 mg/[m.sup.2] of carboplatin, and 1,200 mg/[m.sup.2] of etoposide were administered over the course of 4 days at 4-week intervals.

We treated patients with double-cycle, high-dose chemotherapy followed by PBSCT after conventional chemotherapy or chemoradiotherapy. Our main objective was to examine whether double-cycle, high-dose chemotherapy with ICE can be delivered with acceptable toxicity after conventional chemotherapy or chemoradiotherapy.

MATERIALS AND METHODS

Patients

Patients with histologically confirmed SCLC of any stage were eligible for the study. Eligibility criteria included the following: (1) no previous treatment, including radiotherapy, chemotherapy, and surgery; (2) lesions that could be measured or assessed; (3) age 18 to 65 years; (4) an Eastern Cooperative Oncology Group performance status of 0 or 1; (5) a life expectancy of [greater than or equal to] 12 weeks; (6) a blood count within the normal range, and normal cardiac, hepatic, and renal functions; and (7) Pa[O.sub.2] [greater than or equal to] 70 mm Hg in a sample of arterial blood in patients with LD. As for renal function, a serum creatinine level < 1.5 mg/dL and a creatinine clearance [greater than or equal to] 60 mL/min were required at entry, immediately before the first high-dose chemotherapy, and immediately before the second high-dose chemotherapy. This study was approved by our institutional review board, and all patients provided their informed consent before enrollment.

Before study entry, all patients underwent staging investigations, including physical examination, chest radiography, CT of the chest and abdomen, brain MRI, bone scintigraphy, full blood count, electrolyte measurements, liver and renal function tests, and fiberoptic bronchoscopy with biopsy. LD was defined as tumor confined to one hemithorax with or without ipsilateral supraclavicular lymphadenopathy. All other patients were defined as having ED.

Treatment

Induction Therapy and PBSC Collection: The treatment scheme is outlined in Figure 1. Patients initially received two cycles of chemotherapy with PE at 3-week intervals. Cisplatin was administered at a dose of 80 my/[m.sup.2] on day 1. Etoposide was administered at a dose of 100 mg/[m.sup.2] on days 1, 2, and 3. Four weeks after the first two cycles of PE, patients were administered 300 mg/[m.sup.2] of etoposide IV on days 1 to 5, followed by granulocyte colony-stimulating factor (G-CSF), 50 [micro]g/[m.sup.2]/d subcutaneously, to mobilize stem cells into the blood. PBSCs were collected by leukapheresis and cryopreserved. Collection was performed up to three times, until sufficient PBSCs were obtained to support two cycles of high-dose chemotherapy. Consecutively, two additional cycles of PE were administered at 3-week intervals.

[FIGURE 1 OMITTED]

High-Dose Therapy Followed by PBSCT: Three weeks after the last two cycles of PE, patients received high-dose ICE therapy. Ifosfamide was administered at 2.5 g/[m.sup.2]/d as a 3-h IV infusion for 4 days. Carboplatin was administered at 300 mg/[m.sup.2]/d as a 6-h IV infusion for 4 days. Etoposide was administered at 200 mg/[m.sup.2]/d as a continuous infusion for 5 days. Mesna, 1,000 mg/[m.sup.2], was administered as a 1-h IV infusion 1 h before ifosfamide administration. Subsequently, mesna was administered at 4,000 mg/[m.sup.2]/d as a continuous infusion for 4 days. Forty-eight hours after the end of chemotherapy, PBSCs were reinfused and G-CSF was introduced at 50 [micro]g/kg/d, administered subcutaneously until hematopoietic recovery. If sufficient PBSCs were obtained to allow transplantation twice, a second course of high-dose ICE therapy was administered 3 to 4 months after the first course.

Radiotherapy: Patients with LD received a total dose of 50 Gy of thoracic radiotherapy concurrently with the last two cycles of PE (2.0 Gy/d in 25 fractions over a period of 5 weeks). The target volume for thoracic radiotherapy included the gross tumor, as defined by the chest CT scan, and the bilateral mediastinal and ipsilateral hilar lymph nodes. If involved by tumor, the supraclavicular lymph nodes were also irradiated. The clinically determined volume was expanded by a 1.5-cm margin. After the first course of high-dose ICE therapy, prophylactic whole-brain irradiation was administered to patients with a CR or near-CR (nCR) from the year 2000. The brain irradiation consisted of 15 doses of 2.0 Gy over the course of 3 weeks, for a total dose of 30 Gy.

Assessment of Toxicity, Response, and Survival

General toxicity was classified according to the National Cancer Institute common toxicity criteria. Response was determined by clinical examination and chest CT. Bronchoscopy with biopsy was performed. Response was defined as follows: (1) CR, if all target lesions disappeared; (2) nCR, if there was a > 90% reduction in the sum of the longest diameters of target lesions with persistent radiographic abnormalities; and (3) partial response (PR), if the reduction was at least a 30% reduction in the sum of the longest diameters of target lesions. Response had to be present for 4 weeks with no lesion recurrence. Stable disease was defined as a < 30% reduction or < 9.0% increase in the sum of the longest diameters of target lesions for [greater than or equal to] 8 weeks. Restaging was done by chest CT, brain MRI, and bronchoscopy with biopsy in all patients three times: just before the first high-dose ICE therapy, 4 weeks after the first high-dose ICE therapy, and 4 weeks after the second high-dose ICE therapy. In addition, abdomen CT, bone scintigraphy, or both were performed in patients with distant metastasis to evaluate metastatic lesions. Overall survival was calculated from the first day of chemotherapy to the day of death using the Kaplan-Meier method. The patients were followed up for at least 5 years from the start of treatment.

RESULTS

Patients Characteristics

Twenty patients were enrolled at our hospital between 1995 and 2003. Two patients were excluded: one patient declined to receive high-dose chemotherapy, and the other patient had a cerebral infarction during the second cycle of PE. The clinical characteristics of the patients data are shown in Table 1. The median age was 58 years (range, 46 to 65 years). Fourteen patients were men. Eleven patients had LD, and 7 patients had ED.

Leukaphereses and PBSC Collection

After administration of etoposide, 1,500 mg/[m.sup.2] IV, over the course of 5 days, G-CSF was administered at 50 [micro]g/kg/d for a median of 12 days (range, 10 to 14 days). The interval from day 1 of mobilization to the start of leukapheresis was 17 days (range, 15 to 19 days). The median number of leukaphereses performed per patient was two (range, one to three procedures). The median number of CD34+ cells collected was 13.11 x [10.sup.6]/kg (range, 3.55 to 80.60 x [10.sup.6]/kg). Grade 4 neutropenia occurred in all patients for a median of 5 days (range, 4 to 6 days), and five patients had febrile neutropenia.

Hematopoietic Recovery

Among the 18 patients, 15 patients received high-dose ICE therapy followed by PBSCT twice. The other three patients did not receive the second course of high-dose ICE: two patients had grade 3 renal toxicity during the first course of high-dose ICE, and the other patient lacked sufficient cells for the second PBSCT. The median number of CD34+ cells reinfused was 3.60 x [10.sup.6]/kg (range, 1.4 to 15.6 x [10.sup.6]/kg) in the first PBSCT and 2.54 x [10.sup.6]/kg (range, 1.50 to 19.2 x [10.sup.6]/kg) in the second PBSCT. The median time from PBSCT to a neutrophil count [greater than or equal to] 500/[micro]L was 10 days (range, 8 to 14 days) in the first PBSCT and 10 days (range, 8 to 13 days) in the second PBSCT. The median time from PBSCT to a platelet count [greater than or equal to] 50,000/[micro]L was 14.5 days (range, 11 to 28 days) in the first PBSCT and 15 days (range, 12 to 24 days) in the second PBSCT. There were no differences in hematopoietic recovery between the first and second PBSCT.

Nonhematologic Toxicity

A total of 33 cycles of high-dose ICE therapy were analyzed for nonhematologic toxicity (Table 2). Grade 3 diarrhea occurred in one cycle, and grade 3 renal toxicity in two cycles. Infectious complications occurred in 16 cycles.

Response and Survival

Among the 18 patients evaluated, the response to treatment at study end point was CR in 13 patients, nCR in 2 patients, and PR in 2 patients. PE or PE with concurrent radiotherapy led to a CR or nCR in 10 patients and a PR in 5 patients. Two of the patients with PR had a CR after high-dose ICE therapy. The 2-year and 5-year survival rates were 72% and 55% in patients with LD, and 43% and 0% in those with ED, respectively (Fig 2).

[FIGURE 2 OMITTED]

DISCUSSION

In the 1980s, high-dose chemotherapy with ABMT was administered as first-line treatment. The rationale of early intensification schedules was to intensify therapy for all patients, while avoiding drug resistance induced by previous chemotherapy. (22-24) In the late 1990s, multicyclic high-dose chemotherapy supported by PBSCT or whole blood transfusions was used as an early intensification strategy. (16,24) This approach resulted in high rates of complete remission (56% to 67%) among patients with LD. Late-intensification schedules, in which conventional chemotherapy is initially administered to debulk the tumor, followed by high-dose chemotherapy to promote consolidation, were also tried. In the 1980s, ABMT was used as hematopoietic support (25-28); PB-SCT has been used more recently. (17,18) High-dose chemotherapy appears to be more toxic when used for late intensification than for early intensification, with much higher treatment-related mortality associated with the former than the latter. However, high-dose chemotherapy can be administered more safely and effectively owing to improvements in supportive care, including the availability of hematopoietic growth factors and improved techniques for PBSCT. We initially administered four cycles of PE with concurrent radiotherapy to patients with LD and four cycles of PE alone to those with ED to derive the full effect of conventional chemotherapy or chemoradiotherapy and decrease tumor volume. The patients then received double-cycle, high-dose ICE therapy. There was no mortality related to high-dose ICE therapy followed by PBSCT. Among the 18 patients evaluated, 10 patients had a CR or nCR after PE or PE with concurrent radiotherapy. Among the five patients with a PR, two had a CR after high-dose ICE therapy. The 2-year and 5-year survival rates were 72% and 55% in patients with LD and 43% and 0% in patients with ED, respectively.

In our study, double-cycle, high-dose ICE therapy was administered according to a late-intensification protocol. Our initial concern was that toxicity might cumulatively increase during the second course of high-dose ICE therapy, but this concern proved to be unfounded. The doses of ICE were similar to those used in previous studies. (16,18) Among 33 administered cycles of high-dose ICE therapy, reversible severe diarrhea occurred in one cycle and reversible severe renal toxicity occurred in two cycles. The median number of days from PBSCT to a neutrophil count [greater than or equal to] 500/[micro]L was 10 days in both the first and second PBSCTs, and the median number of days from PBSCT to a platelet count [greater than or equal to] 50,000/[micro]L was 14.5 days in the first PBSCT and 15 days in the second PBSCT. The first PBSCT and second PBSCT thus had similar times to hematopoietic recovery.

The schedules and dosages of ICE regimens have differed considerably among clinical trials. Although unequivocal conclusions cannot be made, several trends have emerged. First, ICE (16,17) regimens are apparently associated with better results of survival. Our results support a good outcome in terms of survival as well as response. Second, ICE regimens permit double cycles of intensification, without a cumulative increase in toxicity, even after conventional chemotherapy or chemoradiotherapy. We conclude that sequential, double-cycle, high-dose ICE therapy is tolerable and feasible in patients with SCLC, including those who have already received full courses of conventional chemotherapy or chemoradiotherapy.

Manuscript received January 30, 2005; revision accepted April 26, 2005.

REFERENCES

(1) Roth BJ, Johnson DH, Einhorn LH, et al. Randomized study of cyclophosphamide, doxorubicin and vincristine versus etoposide and cisplatin versus alternation of these two regimens in extensive small-cell lung cancer: a phase III trial of the Southeastern Cancer Study Group. J Clin Oncol 1992; 10: 282-291

(2) Fukuoka M, Furuse K, Saijo N, et al. Randomized trial of cyclophosphamide, doxorubicin and vincristine versus cisplatin and etoposide versus alternation of these regimens in small-cell lung cancer. J Natl Cancer Inst 1991; 83:855-861

(3) Evans WK, Feld R, Murray N, et al. Superiority of alternating non-cross-resistant chemotherapy in extensive small cell lung cancer: a multicenter, randomized clinical trial by the National Cancer Institute of Canada. Ann Intern Med 1987; 107:451-458

(4) Murray N, Shah A, Osoba D, et al. Intensive weekly chemotherapy for the treatment of extensive-stage small-cell lung cancer. J Clin Oncol 1991; 83:855-861

(5) Furuse K, Fukuoka M, Nishiwaki Y, et al. Phase III study of granulocyte colony-stimulating factor versus standard chemotherapy in extensive-disease small-cell lung cancer. J Clin Oncol 1998; 16:2126-2132

(6) Murray N, Livingston RB, Shephers FA, et al. Randomized study of CODE versus alternating CAV/EP for extensive-stage small-cell lung cancer: an intergroup study of the National Cancer Institute of Canada Clinical Trials Group and the Southwest Oncology Group. J Clin Oncol 1999; 17:2300-2308

(7) Arriagada R, Le Chevalier T, Pierre Pignon J, et al. Initial chemotherapeutic doses and survival in patients with limited small-cell lung cancer, N Engl J Med 1993; 25:1848-1852

(8) Ihde DC, Mulshine JL, Kramer BS, et al. Prospective randomized comparison of high-dose and standard-dose etoposide and cisplatin chemotherapy in patients with extensive-stage small-cell lung cancer. J Clin Oncol 1994; 12:2022-2034

(9) Steward WP, von Pawel J, Gatzemeier U, et al. Effect of granulocyte colony-stimulating factor and dose-intensification of V-ICE chemotherapy in small-cell lung cancer: a prospective randomized study 300 patients. J Clin Oncol 1998; 16:642-650

(10) Thatcher N, Girling DJ, Hopwood P, et al. Improving survival without reducing quality of life in small-cell lung cancer patients by increasing the dose-intensity of chemotherapy with granulocyte colony-stimulating factor support: results of a BMJ Research Council multicenter randomized trial. J Clin Oncol 2000; 18:395-404

(11) Bunn PA Jr., Crowley K, Kelly K, et al. Chemoradiotherapy with/ without granulocyte-macrophage colony-stimulating factor in the treatment of limited stage small cell lung cancer: a prospective phase III randomized study of the Southwest Oncology Group. J Clin Oncol 1995; 13:1632-1641

(12) Sculier JP, Paesmans M, Lecomte J, et al. A three-arm phase III randomized trial assessing, in patients with extensive-disease small-cell lung cancer, accelerated chemotherapy with support of haematological growth factor or oral antibiotics. Br J Cancer 2001; 85:1444-1451

(13) Pasini F, Pelosi G, Manzoni G, et al. High-dose chemotherapy in small cell lung cancer. Tumori 2002; 88:179-186

(14) Elias AD, Ibrahim J, Skavin AT, et al. Dose-intensive therapy for limited-stage small-cell lung cancer: long-term outcome. J Clin Oncol 1999; 17:1175-1184

(15) Humblet Y, Symann M, Bosly A, et al. Late intensification chemotherapy with autologous bone marrow transplantation in selected small-cell carcinoma of the lung: a randomized study. J Clin Oncol 1987; 5:1864-1873

(16) Leyvraz S, Percy L, Rosti A, et al. Multiple courses of high-dose ifosfamide, carboplatin, and etoposide with peripheral-blood progenitor cells and filgrastim for small-cell lung cancer: a feasibility study by the European Group for blood and marrow transplantation. J Clin Oncol 1999; 17:3531-3539

(17) Fetscher S, Brugger W, Engelhardt R, et al. Dose intense therapy with etoposide, ifosfamide, cisplatin, and epirubicin (VIP-E) in 100 consecutive patients with limited- and extensive-disease small-cell lung cancer. Ann Oncol 1997; 8:49-56

(18) Bessho A, Ueoka H, Kiura K, et al. High-dose ifosfamide, carboplatin and etoposide with autologous peripheral blood progenitor cell transplantation for small-cell lung cancer. Anticancer Res 1999; 19:693-698

(19) Van de Velde H, Bosquee L, Weynants P, et al. Moderate dose-escalation of combination chemotherapy with concomitant thoracic radiotherapy in limited disease small cell lung cancer: prolonged intrathoracic tumour control and high central nervous system relapse rate. Ann Oncol 1999; 10: 1051-1057

(20) Elias AD, Ayash LJ, Wheeler C, et al. Phase I study of high-dose ifosfamide, carboplatin, and etoposide with autologous hematopoietic stem cell support. Bone Marrow Transplant 1995; 15:373-379

(21) Fields KK, Elfenbein GJ, Lazarus HM, et al. Maximum-tolerated doses of ifosfamide, carboplatin, and etoposide given over 6 days followed by autologous stem-cell rescue: toxicity profile. J Clin Oncol 1995; 13:323-332

(22) Souhami RI, Finn G, Gregory WM, et al. High-dose cyclophosphamide in small-cell carcinoma of the lung. J Clin Oncol 1985; 3:958-963

(23) Farha P, Spitzer G, Valdivieso M, et al. High-dose chemotherapy and autologous bone marrow transplantation for the treatment of small cell lung carcinoma. Cancer 1983; 52: 1351-1355

(24) Pettengell R, Woll PJ, Thatcher N, et al. Multicyclic dose-intensive chemotherapy supported by sequential reinfusion of hematopoietic progenitors in whole blood. J Clin Oncol 1995; 13:148-156

(25) Klastersky J, Nicaise C, Longeval E, et al. Cisplatin, adriamycin, and etoposide (CAV) for remission induction of small cell bronchogenic carcinoma. Cancer 1982; 50:652-658

(26) Stewart P, Buckner CD, Thomas ED, et al. Intensive chemoradiotherapy with autologous marrow transplantation for small cell carcinoma of the lung. Cancer Treat Rep 1983; 67:1055-1059

(27) Stahel RA, Takvorian RW, Skarin AT, et al. Autologous bone marrow transplantation following high-dose chemotherapy with cyclophosphamide, BCNU and VP-16 in small cell carcinoma of the lung and a review of current literature. Eur J Cancer Clin Oncol 1984; 20:1233-1238

(28) Ihde DC, Deisseroth AB, Lichter AS, et al. Late intensive combined modality therapy followed by autologous bone marrow infusion in extensive-stage small-cell lung cancer. J Clin Oncol 1986; 4:1443-1454

Yoshinobu Iwasaki, MD, PhD; Kazuhiro Nagata, MD; Masaki Nakanishi, MD; Atsushi Natuhara, MD; Yutaka Kubota, MD; Mikio Ueda, MD; Taichiro Arimoto, MD, PhD; and Hiroshi Hara, MD, PhD

* From the Division of Pulmonary Medicine, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan.

Correspondence to: Yoshinobu Iwasaki, MD, PhD, Division of Pulmonary Medicine, Department of Medicine, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji, Kamigyo-ku, Kyoto 602, Japan; e-mail: yiwasaki@koto.kpu-m.ac.jp

COPYRIGHT 2005 American College of Chest Physicians
COPYRIGHT 2005 Gale Group

Return to Carboplatin
Home Contact Resources Exchange Links ebay