全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Molecular Targets for the Treatment of Juvenile Myelomonocytic Leukemia

DOI: 10.1155/2012/308252

Full-Text   Cite this paper   Add to My Lib

Abstract:

Significant advances in our understanding of the genetic defects and the pathogenesis of juvenile myelomonocytic leukemia (JMML) have been achieved in the last several years. The information gathered tremendously helps us in designing molecular targeted therapies for this otherwise fatal disease. Various approaches are being investigated to target defective pathways/molecules in this disease. However, effective therapy is still lacking. Development of specific target-based drugs for JMML remains a big challenge and represents a promising direction in this field. 1. Juvenile Myelomonocytic Leukemia (JMML) and Current Clinical Standard of Care Juvenile myelomonocytic leukemia (JMML) is a rare hematologic malignancy of early childhood with high mortality. It represents 2% to 3% of all pediatric leukemias [1, 2], and its incidence is approximately 0.6 per million children per year [3]. Clinically, patients often present with pallor, failure to thrive, decreased appetite, irritability, dry cough, tachypnea, skin rashes, and diarrhea and are found to have lymphadenopathy and hepatosplenomegaly on examination [4–8]. JMML is characterized by leukocytosis with prominent monocytosis, thrombocytopenia, elevation of fetal hemoglobin (HbF), and hypersensitivity of hematopoietic progenitors to granulocyte-macrophage colony-stimulating factor (GM-CSF) [4–8]. Prior to the revision in 2008, JMML was diagnosed based on the following criteria: presence of peripheral blood monocytosis (>1000/μL); less than 20% blasts in the bone marrow; absence of Philadelphia (Ph) chromosome or BCR-ABL fusion gene AND at least two of the following criteria: increased HbF levels; presence of immature myeloid precursors in the peripheral blood; white blood cell count >10,000/μL; GM-CSF hypersensitivity of myeloid progenitors in vitro [5, 9]. In 2008, the JMML diagnostic criteria were revised to account for the molecular genetic abnormalities that were identified in this disease [5]. The natural course of JMML is rapidly fatal with 80% of patients surviving less than three years [10]. Low platelet count, age at diagnosis older than 2 years, and high HbF percentage have been shown to correlate with poor outcome [11]. Allogeneic hematopoietic stem cell transplantation (HSCT) is currently the only curative treatment for JMML, but controversy exists in identifying the patients that need to proceed to transplant immediately versus those that can be observed for a longer time. Patients with Noonan syndrome often develop a JMML-like myeloproliferative disorder that may resolve spontaneously within

References

[1]  M. H. Freedman, Z. Estrov, and H. S. L. Chan, “Juvenile chronic myelogenous leukemia,” American Journal of Pediatric Hematology/Oncology, vol. 10, no. 3, pp. 261–267, 1988.
[2]  C. M. Niemeyer, M. Arico, G. Basso, et al., “Chronic myelomonocytic leukemia in childhood: a retrospective analysis of 110 cases. European Working Group on Myelodysplastic Syndromes in Childhood (EWOG-MDS),” Blood, vol. 89, no. 10, pp. 3534–3543, 1997.
[3]  A. Manabe, J. Okamura, K. Yumura-Yagi et al., “Allogeneic hematopoietic stem cell transplantation for 27 children with juvenile myelomonocytic leukemia diagnosed based on the criteria of the international JMML working group,” Leukemia, vol. 16, no. 4, pp. 645–649, 2002.
[4]  H. Castro-Malaspina, G. Schaison, and S. Passe, “Subacute and chronic myelomonocytic leukemia in children (juvenile CML). Clinical and hematologic observations and identification of prognostic factors,” Cancer, vol. 54, no. 4, pp. 675–686, 1984.
[5]  R. J. Chan, T. Cooper, C. P. Kratz, B. Weiss, and M. L. Loh, “Juvenile myelomonocytic leukemia: a report from the 2nd International JMML Symposium,” Leukemia Research, vol. 33, no. 3, pp. 355–362, 2009.
[6]  P. D. Emanuel, “Juvenile myelomonocytic leukemia and chronic myelomonocytic leukemia,” Leukemia, vol. 22, no. 7, pp. 1335–1342, 2008.
[7]  P. D. Emanuel, “Myelodysplasia and myeloproliferative disorders in childhood: an update,” The British Journal of Haematology, vol. 105, no. 4, pp. 852–863, 1999.
[8]  P. O. Iversen, I. D. Lewis, S. Turczynowicz et al., “Inhibition of granulocyte-macrophage colony-stimulating factor prevents dissemination and induces remission of juvenile myelomonocytic leukemia in engrafted immunodeficient mice,” Blood, vol. 90, no. 12, pp. 4910–4917, 1997.
[9]  Y. Kimura, Y. Sugita, R. Seki et al., “Infant juvenile myelomonocytic leukemia (JMML) with rapid infiltration of multiple organs,” Pathology International, vol. 60, no. 4, pp. 333–335, 2010.
[10]  M. L. Loh, “Childhood myelodysplastic syndrome: focus on the approach to diagnosis and treatment of juvenile myelomonocytic leukemia,” Hematology. American Society of Hematology. Education Program, vol. 2010, pp. 357–362, 2010.
[11]  S. J. Passmore, J. M. Chessells, H. Kempski, I. M. Hann, P. A. Brownbill, and C. A. Stiller, “Paediatric myelodysplastic syndromes and juvenile myelomonocytic leukaemia in the UK: a population-based study of incidence and survival,” The British Journal of Haematology, vol. 121, no. 5, pp. 758–767, 2003.
[12]  C. P. Kratz, C. M. Niemeyer, R. P. Castleberry et al., “The mutational spectrum of PTPN11 in juvenile myelomonocytic leukemia and Noonan syndrome/myeloproliferative disease,” Blood, vol. 106, no. 6, pp. 2183–2185, 2005.
[13]  F. Locatelli, P. N?llke, M. Zecca et al., “Hematopoietic stem cell transplantation (HSCT) in children with juvenile myelomonocytic leukemia (JMML): results of the EWOG-MDS/EBMT trial,” Blood, vol. 105, no. 1, pp. 410–419, 2005.
[14]  M. L. Loh, “Recent advances in the pathogenesis and treatment of juvenile myelomonocytic leukaemia,” The British Journal of Haematology, vol. 152, no. 6, pp. 677–687, 2011.
[15]  P. D. Emanuel, L. J. Bates, R. P. Castleberry, R. J. Gualtieri, and K. S. Zuckerman, “Selective hypersensitivity to granulocyte-macrophage colony-stimulating factor by juvenile chronic myeloid leukemia hematopoietic progenitors,” Blood, vol. 77, no. 5, pp. 925–929, 1991.
[16]  R. J. Chan, M. B. Leedy, V. Munugalavadla et al., “Human somatic PTPN11 mutations induce hematopoietic-cell hypersensitivity to granulocyte-macrophage colony-stimulating factor,” Blood, vol. 105, no. 9, pp. 3737–3742, 2005.
[17]  C. Flotho, C. Kratz, and C. M. Niemeyer, “Targetting RAS signalling pathways in juvenile myelomonocytic leukemia,” Current Drug Targets, vol. 8, no. 6, pp. 715–725, 2007.
[18]  M. Tartaglia, C. M. Niemeyer, A. Fragale et al., “Somatic mutations in PTPN11 in juvenile myelomonocytic leukemia, myelodysplastic syndromes and acute myeloid leukemia,” Nature Genetics, vol. 34, no. 2, pp. 148–150, 2003.
[19]  M. L. Loh, M. G. Reynolds, S. Vattikuti et al., “PTPN11 mutations in pediatric patients with acute myeloid leukemia: results from the Children's Cancer Group,” Leukemia, vol. 18, no. 11, pp. 1831–1834, 2004.
[20]  N. Yoshida, H. Yagasaki, Y. Xu et al., “Correlation of clinical features with the mutational status of GM-CSF signaling pathway-related genes in juvenile myelomonocytic leukemia,” Pediatric Research, vol. 65, no. 3, pp. 334–340, 2009.
[21]  M. Adachi, M. Sekiya, T. Miyachi et al., “Molecular cloning of a novel protein-tyrosine phosphatase SH-PTP3 with sequence similarity to the src-homology region 2,” FEBS Letters, vol. 314, no. 3, pp. 335–339, 1992.
[22]  S. Ahmad, D. Banville, Z. Zhao, E. H. Fischer, and S. H. Shen, “A widely expressed human protein-tyrosine phosphatase containing src homology 2 domains,” Proceedings of the National Academy of Sciences of the United States of America, vol. 90, no. 6, pp. 2197–2201, 1993.
[23]  G. S. Feng, C. C. Hui, and T. Pawson, “SH2-containing phosphotyrosine phosphatase as a target of protein-tyrosine kinases,” Science, vol. 259, no. 5101, pp. 1607–1611, 1993.
[24]  R. M. Freeman Jr., J. Plutzky, and B. G. Neel, “Identification of a human src homology 2-containing protein-tyrosine-phosphatase: a putative homolog of Drosophila corkscrew,” Proceedings of the National Academy of Sciences of the United States of America, vol. 89, no. 23, pp. 11239–11243, 1992.
[25]  W. Vogel, R. Lammers, J. Huang, and A. Ullrich, “Activation of a phosphotyrosine phosphatase by tyrosine phosphorylation,” Science, vol. 259, no. 5101, pp. 1611–1614, 1993.
[26]  B. G. Neel, H. Gu, and L. Pao, “The 'Shp'ing news: SH2 domain-containing tyrosine phosphatases in cell signaling,” Trends in Biochemical Sciences, vol. 28, no. 6, pp. 284–293, 2003.
[27]  N. K. Tonks, “Protein tyrosine phosphatases: from genes, to function, to disease,” Nature Reviews Molecular Cell Biology, vol. 7, no. 11, pp. 833–846, 2006.
[28]  D. Xu and C. K. Qu, “Protein tyrosine phosphatases in the JAK/STAT pathway,” Frontiers in Bioscience, vol. 13, no. 13, pp. 4925–4932, 2008.
[29]  M. J. Eck, S. Pluskeyt, T. Trüb, S. C. Harrison, and S. E. Shoelson, “Spatial constraints on the recognition of phosphoproteins by the tandem SH2 domains of the phosphatase SH-PTP2,” Nature, vol. 379, no. 6562, pp. 277–280, 1996.
[30]  P. Hof, S. Pluskey, S. Dhe-Paganon, M. J. Eck, and S. E. Shoelson, “Crystal structure of the tyrosine phosphatase SHP-2,” Cell, vol. 92, no. 4, pp. 441–450, 1998.
[31]  H. Keilhack, F. S. David, M. McGregor, L. C. Cantley, and B. G. Neel, “Diverse biochemical properties of Shp2 mutants: implications for disease phenotypes,” Journal of Biological Chemistry, vol. 280, no. 35, pp. 30984–30993, 2005.
[32]  T. Araki, M. G. Mohi, F. A. Ismat et al., “Mouse model of Noonan syndrome reveals cell type- and gene dosage-dependent effects of Ptpn11 mutation,” Nature Medicine, vol. 10, no. 8, pp. 849–857, 2004.
[33]  A. Fragale, M. Tartaglia, J. Wu, and B. D. Gelb, “Noonan syndrome-associated SHP2/PTPN11 mutants cause EGF-dependent prolonged GAB1 binding and sustained ERK2/MAPK1 activation,” Human Mutation, vol. 23, no. 3, pp. 267–277, 2004.
[34]  W. M. Yu, H. Daino, J. Chen, K. D. Bunting, and C. K. Qu, “Effects of a leukemia-associated gain-of-function mutation of SHP-2 phosphatase on interleukin-3 signaling,” Journal of Biological Chemistry, vol. 281, no. 9, pp. 5426–5434, 2006.
[35]  S. Schubbert, K. Lieuw, S. L. Rowe et al., “Functional analysis of leukemia-associated PTPN11 mutations in primary hematopoietic cells,” Blood, vol. 106, no. 1, pp. 311–317, 2005.
[36]  G. Chan, D. Kalaitzidis, T. Usenko et al., “Leukemogenic Ptpn11 causes fatal myeloproliferative disorder via cell-autonomous effects on multiple stages of hematopoiesis,” Blood, vol. 113, no. 18, pp. 4414–4424, 2009.
[37]  M. G. Mohi, I. R. Williams, C. R. Dearolf et al., “Prognostic, therapeutic, and mechanistic implications of a mouse model of leukemia evoked by Shp2 (PTPN11) mutations,” Cancer Cell, vol. 7, no. 2, pp. 179–191, 2005.
[38]  D. Xu, S. Wang, W. M. Yu et al., “A germline gain-of-function mutation in Ptpn11 (Shp-2) phosphatase induces myeloproliferative disease by aberrant activation of hematopoietic stem cells,” Blood, vol. 116, no. 18, pp. 3611–3621, 2010.
[39]  A. C. de Vries, C. M. Zwaan, and M. M. van den Heuvel-Eibrink, “Molecular basis of juvenile myelomonocytic leukemia,” Haematologica, vol. 95, no. 2, pp. 179–182, 2010.
[40]  C. Flotho, S. Valcamonica, S. Mach-Pascual et al., “RAS mutations and clonality analysis in children with juvenile myelomonocytic leukemia (JMML),” Leukemia, vol. 13, no. 1, pp. 32–37, 1999.
[41]  P. De Filippi, M. Zecca, D. Lisini et al., “Germ-line mutation of the NRAS gene may be responsible for the development of juvenile myelomonocytic leukaemia,” The British Journal of Haematology, vol. 147, no. 5, pp. 706–709, 2009.
[42]  K. M. Shannon, P. O'Connell, G. A. Martin et al., “Loss of the normal NF1 allele from the bone marrow of children with type 1 neurofibromatosis and malignant myeloid disorders,” The New England Journal of Medicine, vol. 330, no. 9, pp. 597–601, 1994.
[43]  A. Theos and B. R. Korf, “Pathophysiology of neurofibromatosis type 1,” Annals of Internal Medicine, vol. 144, no. 11, pp. 842–849, 2006.
[44]  C. Flotho, D. Steinemann, C. G. Mullighan et al., “Genome-wide single-nucleotide polymorphism analysis in juvenile myelomonocytic leukemia identifies uniparental disomy surrounding the NF1 locus in cases associated with neurofibromatosis but not in cases with mutant RAS or PTPN11,” Oncogene, vol. 26, no. 39, pp. 5816–5821, 2007.
[45]  L. E. Side, P. D. Emanuel, B. Taylor et al., “Mutations of the NF1 gene in children with juvenile myelomonocytic leukemia without clinical evidence of neurofibromatosis, type 1,” Blood, vol. 92, no. 1, pp. 267–272, 1998.
[46]  M. Aricò, A. Biondi, and C. H. Pui, “Juvenile myelomonocytic leukemia,” Blood, vol. 90, no. 2, pp. 479–488, 1997.
[47]  M. L. Loh, D. S. Sakai, C. Flotho et al., “Mutations in CBL occur frequently in juvenile myelomonocytic leukemia,” Blood, vol. 114, no. 9, pp. 1859–1863, 2009.
[48]  H. Muramatsu, H. Makishima, A. M. Jankowska et al., “Mutations of an E3 ubiquitin ligase c-Cbl but not TET2 mutations are pathogenic in juvenile myelomonocytic leukemia,” Blood, vol. 115, no. 10, pp. 1969–1975, 2010.
[49]  B. Pérez, F. Mechinaud, C. Galambrun et al., “Germline mutations of the CBL gene define a new genetic syndrome with predisposition to juvenile myelomonocytic leukaemia,” Journal of Medical Genetics, vol. 47, no. 10, pp. 686–691, 2010.
[50]  C. M. Niemeyer, M. W. Kang, D. H. Shin et al., “Germline CBL mutations cause developmental abnormalities and predispose to juvenile myelomonocytic leukemia,” Nature Genetics, vol. 42, no. 9, pp. 794–800, 2010.
[51]  Y. Sugimoto, H. Muramatsu, H. Makishima et al., “Spectrum of molecular defects in juvenile myelomonocytic leukaemia includes ASXL1 mutations,” The British Journal of Haematology, vol. 150, no. 1, pp. 83–87, 2010.
[52]  E. J. Gratias, Y. L. Liu, S. Meleth, R. P. Castleberry, and P. D. Emanuel, “Activating FLT3 mutations are rare in children with juvenile myelomonocytic leukemia,” Pediatric Blood and Cancer, vol. 44, no. 2, pp. 142–146, 2005.
[53]  D. Steinemann, M. Tauscher, I. Praulich, C. M. Niemeyer, C. Flotho, and B. Schlegelberger, “Mutations in the let-7 binding site—a mechanism of RAS activation in juvenile myelomonocytic leukemia?” Haematologica, vol. 95, no. 9, p. 1616, 2010.
[54]  B. Pérez, O. Kosmider, B. Cassinat et al., “Genetic typing of CBL, ASXL1, RUNX1, TET2 and JAK2 in juvenile myelomonocytic leukaemia reveals a genetic profile distinct from chronic myelomonocytic leukaemia,” The British Journal of Haematology, vol. 151, no. 5, pp. 460–468, 2010.
[55]  C. Flotho, C. Batz, H. Hasle et al., “Mutational analysis of SHOC2, a novel gene for Noonan-like syndrome, in JMML,” Blood, vol. 115, no. 4, p. 913, 2010.
[56]  S. Luna-Fineman, K. M. Shannon, S. K. Atwater et al., “Myelodysplastic and myeloproliferative disorders of childhood: a study of 167 patients,” Blood, vol. 93, no. 2, pp. 459–466, 1999.
[57]  J. M. Meck, J. A. Otani-Rosa, R. W. Neuberg, J. A. Welsh, P. N. Mowrey, and A. M. Meloni-Ehrig, “A rare finding of deletion 5q in a child with juvenile myelomonocytic leukemia,” Cancer Genetics and Cytogenetics, vol. 195, no. 2, pp. 192–194, 2009.
[58]  J. D. Grainger, A. M. Will, and R. F. Stevens, “Cultured autografting for juvenile myelomonocytic leukaemia,” The British Journal of Haematology, vol. 117, no. 2, pp. 477–479, 2002.
[59]  S. Tosi, G. Mosna, G. Cazzaniga et al., “Unbalanced t(3;12) in a case of juvenile myelomonocytic leukemia (JMML) results in partial trisomy of 3q as defined by FISH,” Leukemia, vol. 11, no. 9, pp. 1465–1468, 1997.
[60]  P. O. Iversen, P. D. Emanuel, and M. Sioud, “Targeting Raf-1 gene expression by a DNA enzyme inhibits juvenile myelomonocytic leukemia cell growth,” Blood, vol. 99, no. 11, pp. 4147–4153, 2002.
[61]  N. Thompson and J. Lyons, “Recent progress in targeting the Raf/MEK/ERK pathway with inhibitors in cancer drug discovery,” Current Opinion in Pharmacology, vol. 5, no. 4, pp. 350–356, 2005.
[62]  J. F. Lyons, S. Wilhelm, B. Hibner, and G. Bollag, “Discovery of a novel Raf kinase inhibitor,” Endocrine-Related Cancer, vol. 8, no. 3, pp. 219–225, 2001.
[63]  N. Mahgoub, B. R. Taylor, M. Gratiot et al., “In vitro and in vivo effects of a farnesyltransferase inhibitor on Nf1-deficient hematopoietic cells,” Blood, vol. 94, no. 7, pp. 2469–2476, 1999.
[64]  P. D. Emanuel, R. C. Snyder, T. Wiley, B. Gopurala, and R. P. Castleberry, “Inhibition of juvenile myelomonocytic leukemia cell growth in vitro by farnesyltransferase inhibitors,” Blood, vol. 95, no. 2, pp. 639–645, 2000.
[65]  M. E. Balasis, K. D. Forinash, Y. A. Chen et al., “Combination of farnesyltransferase and Akt inhibitors is synergistic in breast cancer cells and causes significant breast tumor regression in ErbB2 transgenic mice,” Clinical Cancer Research, vol. 17, no. 9, pp. 2852–2862, 2011.
[66]  J. E. Lancet, V. H. Duong, E. F. Winton et al., “A phase I clinical-pharmacodynamic study of the farnesyltransferase inhibitor tipifarnib in combination with the proteasome inhibitor bortezomib in advanced acute leukemias,” Clinical Cancer Research, vol. 17, no. 5, pp. 1140–1146, 2011.
[67]  L. Chen, S. S. Sung, M. L. R. Yip et al., “Discovery of a novel Shp2 protein tyrosine phosphatase inhibitor,” Molecular Pharmacology, vol. 70, no. 2, pp. 562–570, 2006.
[68]  W. M. Yu, O. Guvench, A. D. MacKerell, and C. K. Qu, “Identification of small molecular weight inhibitors of Src homology 2 domain-containing tyrosine phosphatase 2 (SHP-2) via in silico database screening combined with experimental assay,” Journal of Medicinal Chemistry, vol. 51, no. 23, pp. 7396–7404, 2008.
[69]  K. Hellmuth, S. Grosskopf, T. L. Ching et al., “Specific inhibitors of the protein tyrosine phosphatase Shp2 identified by high-throughput docking,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 20, pp. 7275–7280, 2008.
[70]  X. Zhang, Y. He, S. Liu et al., “Salicylic acid based small molecule inhibitor for the oncogenic src homology-2 domain containing protein tyrosine phosphatase-2 (SHP2),” Journal of Medicinal Chemistry, vol. 53, no. 6, pp. 2482–2493, 2010.
[71]  M. H. Freedman, A. Cohen, T. Grunberger et al., “Central role of tumour necrosis factor, GM-CSF, and interleukin 1 in the pathogenesis of juvenile chronic myelogenous leukaemia,” The British Journal of Haematology, vol. 80, no. 1, pp. 40–48, 1992.
[72]  P. O. Iversen, R. L. Rodwell, L. Pitcher, K. M. Taylor, and A. F. Lopez, “Inhibition of proliferation and induction of apoptosis in juvenile myelomonocytic leukemic cells by the granulocyte-macrophage colony-stimulating factor analogue E21R,” Blood, vol. 88, no. 7, pp. 2634–2639, 1996.
[73]  P. O. Iversen, D. R. Sorensen, and H. B. Benestad, “Inhibitors of angiogenesis selectively reduce the malignant cell load in rodent models of human myeloid leukemias,” Leukemia, vol. 16, no. 3, pp. 376–381, 2002.
[74]  M. E. Van Meter, E. Díaz-Flores, J. A. Archard et al., “K-RasG12D expression induces hyperproliferation and aberrant signaling in primary hematopoietic stem/progenitor cells,” Blood, vol. 109, no. 9, pp. 3945–3952, 2007.
[75]  J. Zhang, Y. Liu, C. Beard et al., “Expression of oncogenic K-ras from its endogenous promoter leads to a partial block of erythroid differentiation and hyperactivation of cytokine-dependent signaling pathways,” Blood, vol. 109, no. 12, pp. 5238–5241, 2007.
[76]  J. Wang, Y. Liu, Z. Li et al., “Endogenous oncogenic NRAS mutation promotes aberrant GM-CSF signaling in granulocytic/monocytic precursors in a murine model of chronic myelomonocytic leukemia,” Blood, vol. 116, no. 26, pp. 5991–6002, 2010.
[77]  N. Kotecha, N. J. Flores, J. M. Irish et al., “Single-cell profiling identifies aberrant STAT5 activation in myeloid malignancies with specific clinical and biologic correlates,” Cancer Cell, vol. 14, no. 4, pp. 335–343, 2008.
[78]  N. Harir, C. Pecquet, M. Kerenyi et al., “Constitutive activation of Stat5 promotes its cytoplasmic localization and association with PI3-kinase in myeloid leukemias,” Blood, vol. 109, no. 4, pp. 1678–1686, 2007.
[79]  K. Matsuda, A. Shimada, N. Yoshida et al., “Spontaneous improvement of hematologic abnormalities in patients having juvenile myelomonocytic leukemia with specific RAS mutations,” Blood, vol. 109, no. 12, pp. 5477–5480, 2007.
[80]  M. Fukuda, K. Horibe, Y. Miyajima, K. Matsumoto, and M. Nagashima, “Spontaneous remission of juvenile chronic myelomonocytic leukemia in an infant with Noonan syndrome,” Journal of Pediatric Hematology/Oncology, vol. 19, no. 2, pp. 177–179, 1997.
[81]  R. P. Castleberry, P. D. Emanuel, K. S. Zuckerman et al., “A pilot study of isotretinoin in the treatment of juvenile chronic myelogenous leukemia,” The New England Journal of Medicine, vol. 331, no. 25, pp. 1680–1684, 1994.
[82]  S. J. Cohen, L. Ho, S. Ranganathan et al., “Phase II and pharmacodynamic study of the farnesyltransferase inhibitor R115777 as initial therapy in patients with metastatic pancreatic adenocarcinoma,” Journal of Clinical Oncology, vol. 21, no. 7, pp. 1301–1306, 2003.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413