51
|
Dy GK, Haluska P, Adjei AA. Novel pharmacological agents in clinical development for solid tumours. Expert Opin Investig Drugs 2001; 10:2059-88. [PMID: 11772306 DOI: 10.1517/13543784.10.12.2059] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
For decades, cancer therapy has focused on DNA-directed mechanisms of cytotoxicity, utilising agents with limited efficacy and significant toxicity. Recent advances in tumour biology have elucidated the molecular pathways implicated in the pathogenesis and progression of cancers and have resulted in the discovery of a variety of novel molecular targets for therapeutic intervention. Promising novel agents targeting signal transduction pathways, cell cycle regulation, angiogenesis and apoptosis are in clinical testing and are discussed in this review.
Collapse
Affiliation(s)
- G K Dy
- Division of Medical Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | |
Collapse
|
52
|
Mohammad RM, Adsay NV, Philip PA, Pettit GR, Vaitkevicius VK, Sarkar FH. Bryostatin 1 induces differentiation and potentiates the antitumor effect of Auristatin PE in a human pancreatic tumor (PANC-1) xenograft model. Anticancer Drugs 2001; 12:735-40. [PMID: 11593055 DOI: 10.1097/00001813-200110000-00005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Pancreatic cancer has the worst prognosis of all cancers with a dismal 5-year survival rate. Hence, there is a tremendous need for development of new and effective therapy for this tumor. In an earlier study we reported a potent antitumor activity of Auristatin PE (AuriPE) against pancreatic tumor. In addition, we have also reported that bryostatin 1 (bryo1) induces differentiation of leukemia cells, but the effect of bryo1 has not been investigated in pancreatic tumors. This is the first report where we demonstrate that bryo1 induces differentiation and potentiates the antitumor effect of AuriPE in a human pancreatic tumor (PANC-1) xenograft model. A xenograft model was established by injecting the PANC-1 cells s.c. in severe combined immune deficient (SCID) mice. After development of the s.c. tumors, tumors were dissected and small fragments were transplanted in vivo to new SCID mice, with a success rate of 100% and a doubling time of 4.8 days. The SCID mouse xenograft model was used to test the in vivo differentiation effect of bryo1 and its efficacy when given alone or in combination with AuriPE. Sections from paraffin-embedded tumors excised from untreated (control) SCID mice revealed typical poorly differentiated adenocarcinoma of the pancreas. Interestingly, sections of s.c. tumors taken from bryo1-treated mice revealed carcinomas that were much lower grade and less aggressive, and displayed prominent squamous and glandular differentiation. In this study, the tumor growth inhibition (T/C), activity score and cure rate for bryo1, AuriPE and bryo1+AuriPE were 80%, (+) and 0/4; 0.0%, (++++) and 3/5; and 0.0%, (++++) and 3/4, respectively. Mice treated with either AuriPE or bryo1+AuriPE were free of tumors for more than 150 days and were considered cured. The use of bryo1 as a novel differentiating agent and its combination with AuriPE should be further explored for the treatment of adenocarcinoma of the pancreas.
Collapse
Affiliation(s)
- R M Mohammad
- Division of Hematology and Oncology, Department of Internal Medicine, Wayne State University School of Medicine, Detroit, Ml 48201, USA
| | | | | | | | | | | |
Collapse
|
53
|
Abstract
Patients with chemotherapy-resistant acute myeloid leukaemia are rarely cured by non-allogeneic transplant therapies. Multiple new investigational agents have become available for treatment of these patients and there are few tools to permit rational drug and clinical trial selection. In this review, we describe the chemical and biological properties of some of these agents and some of their initial clinical activity to date. The selected agents react with either cell surface molecules or signal pathway intermediates and include antibody and antibody conjugates to CD33 and CD45, a fusion protein directed to the granulocyte-macrophage colony-stimulating factor receptor, an anti-sense oligonucleotide to Bcl2, a farnesyl transferase inhibitor, and a protein kinase C agonist/inhibitor. The challenge for the next decade will be how to select patients for particular molecularly targeted therapeutics and how to combine these agents.
Collapse
Affiliation(s)
- A E Frankel
- Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | | | |
Collapse
|
54
|
Yoo J, Nichols A, Song JC, Mun EC, Matthews JB. Protein kinase epsilon dampens the secretory response of model intestinal epithelia during ischemia. Surgery 2001; 130:310-8. [PMID: 11490365 DOI: 10.1067/msy.2001.116034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND Luminal fluid sequestration and diarrhea are early manifestations of mesenteric ischemia. This can be modeled in vitro with the use of T84 intestinal epithelia, where ischemia induces Cl(-) secretion with adenosine-mediated autocrine feedback. Protein kinase C (PKC) regulates epithelial transport and, in some organ systems, is involved in the response to ischemic stress. The purpose of this study was to define the role of PKC on epithelial transport during ischemia. METHODS By voltage-current clamp, short-circuit current (Isc) equals Cl(-) secretion. Ischemic conditions were simulated with the use of a well-established chemical hypoxia protocol. RESULTS Chemical hypoxia briskly activated Isc. Gö6850, an antagonist of novel and conventional PKC isoforms, markedly enhanced the ischemia-induced Isc response, although Gö6976 (which inhibits only conventional isoforms) had no effect. Rottlerin, a specific inhibitor of PKC delta, did not attenuate ischemic Isc. Both phorbol 12-myristate, 13-acetate and bryostatin-1, which selectively activate PKC epsilon in T84 cells, markedly attenuated the Isc response to ischemia. Both agents also inhibited the Isc response to exogenous adenosine. CONCLUSIONS PKC (likely the novel epsilon isoform) in intestinal epithelia modulates ischemia-induced alterations in ion transport. Inhibition of PKC epsilon exaggerates the secretory response that is induced by ischemia and by authentic adenosine; conversely, augmented activation of PKC epsilon inhibits secretion. Manipulation of PKC epsilon could limit luminal fluid sequestration during mesenteric ischemia.
Collapse
Affiliation(s)
- J Yoo
- Beth Israel Deaconess Medical Center, Division of General and Gastrointestinal Surgery, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | |
Collapse
|
55
|
Galmarini CM, Mackey JR, Dumontet C. Nucleoside analogues: mechanisms of drug resistance and reversal strategies. Leukemia 2001; 15:875-90. [PMID: 11417472 DOI: 10.1038/sj.leu.2402114] [Citation(s) in RCA: 354] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nucleoside analogues (NA) are essential components of AML induction therapy (cytosine arabinoside), effective treatments of lymphoproliferative disorders (fludarabine, cladribine) and are also used in the treatment of some solid tumors (gemcitabine). These important compounds share some general common characteristics, namely in terms of requiring transport by specific membrane transporters, metabolism and interaction with intracellular targets. However, these compounds differ in regard to the types of transporters that most efficiently transport a given compound, and their preferential interaction with certain targets which may explain why some compounds are more effective against rapidly proliferating tumors and others on neoplasia with a more protracted evolution. In this review, we analyze the available data concerning mechanisms of action of and resistance to NA, with particular emphasis on recent advances in the characterization of nucleoside transporters and on the potential role of activating or inactivating enzymes in the induction of clinical resistance to these compounds. We performed an extensive search of published in vitro and clinical data in which the levels of expression of nucleoside-activating or inactivating enzymes have been correlated with tumor response or patient outcome. Strategies aiming to increase the intracellular concentrations of active compounds are presented.
Collapse
Affiliation(s)
- C M Galmarini
- Unité INSERM 453, Laboratoire de Cytologie Analytique, Faculté de Médécine Rockefeller, Lyon, France
| | | | | |
Collapse
|
56
|
Abstract
The non-Hodgkin lymphomas (NHL) are characterized by initial responsiveness to a variety of chemotherapeutic regimens. Nevertheless, most patients progress and die from their disease. A number of new agents with unique mechanisms of action are in clinical development. Agents that are currently considered to be the most promising include those that induce apoptosis; those that interfere with cell cycling, tumor-associated angiogenesis, farnesylation of the Ras gene, and histone deacetylase; and those that inhibit the proteasome, among others. Increasing insights into the differences between tumors and among patients will lead to more individualized therapeutic strategies using agents directed at specific genetic and immunologic targets. More rapid accrual to high-quality clinical studies will facilitate dissemination of new agents to patients and lead to an increased cure rate for NHL.
Collapse
Affiliation(s)
- B D Cheson
- National Cancer Institute, Executive Plaza North, Room 741, Bethesda, MD 20892, USA.
| |
Collapse
|
57
|
Leszczyniecka M, Roberts T, Dent P, Grant S, Fisher PB. Differentiation therapy of human cancer: basic science and clinical applications. Pharmacol Ther 2001; 90:105-56. [PMID: 11578655 DOI: 10.1016/s0163-7258(01)00132-2] [Citation(s) in RCA: 211] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Current cancer therapies are highly toxic and often nonspecific. A potentially less toxic approach to treating this prevalent disease employs agents that modify cancer cell differentiation, termed 'differentiation therapy.' This approach is based on the tacit assumption that many neoplastic cell types exhibit reversible defects in differentiation, which upon appropriate treatment, results in tumor reprogramming and a concomitant loss in proliferative capacity and induction of terminal differentiation or apoptosis (programmed cell death). Laboratory studies that focus on elucidating mechanisms of action are demonstrating the effectiveness of 'differentiation therapy,' which is now beginning to show translational promise in the clinical setting.
Collapse
Affiliation(s)
- M Leszczyniecka
- Department of Urology, Herbert Irving Comprehensive Cancer Center, Columbia University, College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
58
|
Sekine I, Saijo N. Growth-Stimulating Pathways in Lung Cancer: Implications for Targets of Therapy. Clin Lung Cancer 2001; 2:299-306; discussion 307. [PMID: 14720364 DOI: 10.3816/clc.2001.n.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Growth-stimulating pathways activated independently of their normal tissue environment are critical to the carcinogenesis and progression of lung cancer. These pathways are comprised of extracellular growth factors; their specific receptors on the cellular membrane; signal transduction cascades in the cytosol; and target molecules, including cytoskeletal proteins, metabolic regulators, and transcription factors in the nucleus. Growth factors can be divided into two groups based on their receptors: G-protein-coupled receptors and receptor tyrosine kinases. Growth factors induce clonal expansion of lung cancer cells by autocrine and/or paracrine mechanisms. Signal transduction cascades form an extremely large and complicated network with cross-talk connections. Ras, phosphatidylinositol-3-OH kinase, and phospholipase C are three key regulators involved in the network. Recent progress in our understanding of the oncoproteins functioning in the pathways has led to the development of novel therapeutic agents. Some of the most exciting results have been obtained with inhibitors of receptor tyrosine kinases. Phase I studies of epidermal growth factor-receptor inhibitors demonstrate objective responses without severe toxicity as single agents in patients with non-small-cell lung cancer refractory to conventional chemotherapy. This new strategy might lead to breakthroughs in the treatment of lung cancer with distant metastases not curable by conventional chemotherapy alone.
Collapse
Affiliation(s)
- I Sekine
- Internal Medicine and Thoracic Oncology Division, National Cancer Center Hospital, Tokyo, Japan.
| | | |
Collapse
|
59
|
Abstract
Bryostatin 1 is a naturally occurring macrocyclic lactone with promising antitumour and immunomodulatory function in preclinical and phase I clinical investigations. In this phase II study, 17 patients with progressive non-Hodgkin's lymphoma of indolent type (NHL), previously treated with chemotherapy, received a median of 6 (range 1-9) intravenous infusions of 25 microg/m(2) bryostatin 1 given once weekly over 24 hours. In 14 evaluable patients no responses were seen. Stable disease was attained in one patient for 9 months. The principal toxicities were myalgia and phlebitis. Treatment was discontinued early because of toxicity alone (phlebitis) in 2 patients, toxicity in addition to progressive disease in 3 patients (myalgia and phlebitis n = 2; thrombocytopenia n = 1) and progressive disease in 5 patients. The results fail to demonstrate efficacy of this regimen of bryostatin 1 in the treatment of NHL. In light of preclinical data that demonstrate synergy between bryostatin 1 and several cytotoxic agents and cytokines, clinical studies to investigate bryostatin 1 in combination are warranted. We also present data to demonstrate that central venous lines may be used in future studies to avoid phlebitis.
Collapse
|
60
|
Vrana JA, Grant S. Synergistic induction of apoptosis in human leukemia cells (U937) exposed to bryostatin 1 and the proteasome inhibitor lactacystin involves dysregulation of the PKC/MAPK cascade. Blood 2001; 97:2105-14. [PMID: 11264178 DOI: 10.1182/blood.v97.7.2105] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Cotreatment with a minimally toxic concentration of the protein kinase C (PKC) activator (and down-regulator) bryostatin 1 (BRY) induced a marked increase in mitochondrial dysfunction and apoptosis in U937 monocytic leukemia cells exposed to the proteasome inhibitor lactacystin (LC). This effect was blocked by cycloheximide, but not by alpha-amanitin or actinomycin D. Qualitatively similar interactions were observed with other PKC activators (eg, phorbol 12-myristate 13-acetate and mezerein), but not phospholipase C, which does not down-regulate the enzyme. These events were examined in relationship to functional alterations in stress (eg, SAPK, JNK) and survival (eg, MAPK, ERK) signaling pathways. The observations that LC/BRY treatment failed to trigger JNK activation and that cell death was unaffected by a dominant-interfering form of c-JUN (TAM67) or by pretreatment with either curcumin or the p38/RK inhibitor, SB203580, suggested that the SAPK pathway was not involved in potentiation of apoptosis. In marked contrast, perturbations in the PKC/Raf/MAPK pathway played an integral role in LC/BRY-mediated cell death based on evidence that pretreatment of cells with bisindolylmaleimide I, a selective PKC inhibitor, or geldanamycin, a benzoquinone ansamycin, which destabilizes and depletes Raf-1, markedly suppressed apoptosis. Furthermore, ERK phosphorylation was substantially prolonged in LC/BRY-treated cells compared to those exposed to BRY alone, and pretreatment with the highly specific MEK inhibitors, PD98059, U0126, and SL327, opposed ERK activation while protecting cells from LC/BRY-induced lethality. Together, these findings suggest a role for activation and/or dysregulation of the PKC/MAPK cascade in modulation of leukemic cell apoptosis following exposure to the proteasome inhibitor LC. (Blood. 2001;97:2105-2114)
Collapse
Affiliation(s)
- J A Vrana
- Department of Medicine, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA
| | | |
Collapse
|
61
|
Schwartsmann G, Brondani da Rocha A, Berlinck RG, Jimeno J. Marine organisms as a source of new anticancer agents. Lancet Oncol 2001; 2:221-5. [PMID: 11905767 DOI: 10.1016/s1470-2045(00)00292-8] [Citation(s) in RCA: 161] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Various active anticancer agents are derived from plants and terrestrial microorganisms. The isolation of C-nucleosides from the Caribbean sponge, Cryptotheca crypta, four decades ago, provided the basis for the synthesis of cytarabine, the first marine-derived anticancer agent to be developed for clinical use. Cytarabine is currently used in the routine treatment of patients with leukaemia and lymphoma. Gemcitabine, one of its fluorinated derivatives, has also been approved for use in patients with pancreatic, breast, bladder, and non-small-cell lung cancer. Over the past decade, several new experimental anticancer agents derived from marine sources have entered preclinical and clinical trials. This field has expanded significantly as a result of improvements in the technology of deep-sea collection, extraction, and large-scale production through aquaculture and synthesis. In this paper, examples of marine-derived experimental agents that are currently undergoing preclinical and early clinical evaluation are briefly discussed. A summary of the available information on the results of phase I and II trials of agents such as aplidine, ecteinascidin-734 (ET-734), dolastatin 10 and bryostatin 1 is also presented.
Collapse
Affiliation(s)
- G Schwartsmann
- South-American Office for Anticancer Drug Development, Comprehensive Cancer Center, The Lutheran University, Brazil.
| | | | | | | |
Collapse
|
62
|
Bedikian AY, Plager C, Stewart JR, O'Brian CA, Herdman SK, Ross M, Papadopoulos N, Eton O, Ellerhorst J, Smith T. Phase II evaluation of bryostatin-1 in metastatic melanoma. Melanoma Res 2001; 11:183-8. [PMID: 11333129 DOI: 10.1097/00008390-200104000-00014] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In this phase II study we assessed the efficacy of bryostatin-1 (NSC 339555) in metastatic melanoma patients when given intravenously either once a week at a dose of 25 microg/m2 per day over 24 h for 3 weeks or at 40 microg/m2 per day over 72 h every 2 weeks. Treatment courses were repeated every 4 weeks. Patients who had received one prior chemotherapy regimen for advanced melanoma, with or without biotherapy, were randomized to one or the other bryostatin-1 dose schedules until 12 patients were registered to each arm. Because there was one confirmed response among the 12 patients who received the 72 h dose schedule, 25 more patients were added to that arm. No prophylactic medications were given. Objective tumour measurements were used to assess the efficacy of the regimen. The National Cancer Institutes common toxicity criteria were used to grade reactions. In total, 49 patients with metastatic melanoma, none having symptomatic brain metastasis, were studied. Of these, 12 patients received the 24 h bryostatin-1 regimen, while the remaining 37 received the 72 h regimen. One patient receiving the 72 h regimen had a partial response lasting over 7 months. Muscle pain occurred in over 90% of the patients and was the dose-limiting side effect of the 72 h regimen. Grade 3/4 nausea and vomiting were more common on the 24 h regimen than on the 72 h one (35% versus 5% of patients). There was no therapy-related thrombocytopenia. Neutropenia was mild and mainly limited to patients receiving the 72 h regimen. Bryostatin-1 has limited activity against melanoma when given by 72 h intravenous infusion.
Collapse
Affiliation(s)
- A Y Bedikian
- Department of Melanoma/Sarcoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Affiliation(s)
- G Schwartsmann
- South-American Office for Anticancer Drug Development (SOAD), Comprehensive Cancer Centre (CINCAN), Lutheran University (ULBRA), Porto Alegre, Brazil
| |
Collapse
|
64
|
Abstract
PKC isoenzymes were found to be involved in proliferation, antitumor drug resistance and apoptosis. Therefore, it has been tried to exploit PKC as a target for antitumor treatment. PKC alpha activity was found to be elevated, for example, in breast cancers and malignant gliomas, whereas it seems to be underexpressed in many colon cancers. So it can be expected that inhibition of PKC activity will not show similar antitumor activity in all tumors. In some tumors it seems to be essential to inhibit PKC to reduce growth. However, for inhibition of tumor proliferation it may be an advantage to induce apoptosis. In this case an activation of PKC delta should be achieved. The situation is complicated by the facts that bryostatin leads to the activation of PKC and later to a downmodulation and that the PKC inhibitors available to date are not specific for one PKC isoenzyme. For these reasons, PKC modulation led to many contradicting results. Despite these problems, PKC modulators such as miltefosine, bryostatin, safingol, CGP41251 and UCN-01 are used in the clinic or are in clinical evaluation. The question is whether PKC is the major or the only target of these compounds, because they also interfere with other targets. PKC may also be involved in apoptosis. Oncogenes and growth factors can induce cell proliferation and cell survival, however, they can also induce apoptosis, depending on the cell type or conditions in which the cells or grown. PKC participates in these signalling pathways and cross-talks. Induction of apoptosis is also dependent on many additional factors, such as p53, bcl-2, mdm2, etc. Therefore, there are also many contradicting results on PKC modulation of apoptosis. Similar controversial data have been reported about MDR1-mediated multidrug resistance. At present it seems that PKC inhibition alone without direct interaction with PGP will not lead to successful reversal of PGP-mediated drug efflux. One possibility to improve chemotherapy would be to combine established antitumor drugs with modulators of PKC. However, here also very contrasting results were obtained. Many indicate that inhibition, others, that activation of PKC enhances the antiproliferative activity of anticancer drugs. The problem is that the exact functions of the different PKC isoenzymes are not clear at present. So further investigations into the role of PKC isoenzymes in the complex and interacting signalling pathways are essential. It is a major challenge in the future to reveal whether modulation of PKC can be used for the improvement of cancer therapy.
Collapse
Affiliation(s)
- J Hofmann
- Institute of Medical Chemistry and Biochemistry, University of Innsbruck, A-6020 Innsbruck, Austria
| |
Collapse
|
65
|
Blackhall FH, Ranson M, Radford JA, Hancock BW, Soukop M, McGown AT, Robbins A, Halbert G, Jayson GC. A phase II trial of bryostatin 1 in patients with non-Hodgkin's lymphoma. Br J Cancer 2001; 84:465-9. [PMID: 11263437 PMCID: PMC2363763 DOI: 10.1054/bjoc.2000.1624] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Bryostatin 1 is a naturally occurring macrocyclic lactone with promising antitumour and immunomodulatory function in preclinical and phase I clinical investigations. In this phase II study, 17 patients with progressive non-Hodgkin's lymphoma of indolent type (NHL), previously treated with chemotherapy, received a median of 6 (range 1-9) intravenous infusions of 25 microg/m(2) bryostatin 1 given once weekly over 24 hours. In 14 evaluable patients no responses were seen. Stable disease was attained in one patient for 9 months. The principal toxicities were myalgia and phlebitis. Treatment was discontinued early because of toxicity alone (phlebitis) in 2 patients, toxicity in addition to progressive disease in 3 patients (myalgia and phlebitis n = 2; thrombocytopenia n = 1) and progressive disease in 5 patients. The results fail to demonstrate efficacy of this regimen of bryostatin 1 in the treatment of NHL. In light of preclinical data that demonstrate synergy between bryostatin 1 and several cytotoxic agents and cytokines, clinical studies to investigate bryostatin 1 in combination are warranted. We also present data to demonstrate that central venous lines may be used in future studies to avoid phlebitis.
Collapse
Affiliation(s)
- F H Blackhall
- CRC Department of Medical Oncology, Christie Hospital NHS Trust, Manchester, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Nuijen B, Bouma M, Manada C, Jimeno JM, Schellens JH, Bult A, Beijnen JH. Pharmaceutical development of anticancer agents derived from marine sources. Anticancer Drugs 2000; 11:793-811. [PMID: 11142687 DOI: 10.1097/00001813-200011000-00003] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The marine ecosystem is more and more acknowledged as a source of potential anticancer agents. After the identification of a potential substance several hurdles have to be overcome before a marine candidate can enter the clinic. Amongst these are the establishment of a method which ensures sufficient supply and, which is the focus of this review, the development of a clinically useful pharmaceutical formulation. General issues with respect to the pharmaceutical development of marine anticancer agents will be discussed, which will be illustrated by highlighting aspects of the pharmaceutical development and clinical use of some representative compounds.
Collapse
Affiliation(s)
- B Nuijen
- Department of Pharmacy and Pharmacology, Slotervaart Hospital/The Netherlands Cancer Institute, Amsterdam.
| | | | | | | | | | | | | |
Collapse
|
67
|
Abstract
Bryostatins are a class of antineoplastic compounds isolated from the bryozoans Bugula neritina. A wide range of scientific research is currently underway, studying different aspects of the bryostatins. In this review we try to summarize the latest findings, including all the topics involved, from marine biology to medicinal chemistry.
Collapse
Affiliation(s)
- R Mutter
- Department of Chemistry, University of Warwick, Coventry, UK.
| | | |
Collapse
|
68
|
Couderc B, Dujols JP, Mokhtari F, Norkowski JL, Slawinski JC, Schlaifer D. The management of adult aggressive non-Hodgkin's lymphomas. Crit Rev Oncol Hematol 2000; 35:33-48. [PMID: 10863150 DOI: 10.1016/s1040-8428(99)00037-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Aggressive non-Hodgkin's lymphona include diffuse large B-cell lymphoma, anaplastic large cell lymphona, and different peripheral T-cell lymphomas. An international prognostic index has been developed including age, serum LDH, performance status, and extranodal involvement. For localized aggressive lymphoma, the preferred treatment is 3-4 CHOP and radiation therapy, with a cure rate of 70-80%. For disseminated aggressive lymphoma, current regimens have a cure rate of less than 40%. Innovative strategies, including dose escalation, autologus stem cell support, new drugs, and immunotherapy are being explored to improve these results.
Collapse
Affiliation(s)
- B Couderc
- Groupe de Radiothérapie et d'Oncologie médicale des Pyrénées (GROP), chemin de l'Ormeau, 65000, Tarbes, France
| | | | | | | | | | | |
Collapse
|
69
|
Damiens E. Molecular events that regulate cell proliferation: an approach for the development of new anticancer drugs. PROGRESS IN CELL CYCLE RESEARCH 2000; 4:219-33. [PMID: 10740828 DOI: 10.1007/978-1-4615-4253-7_19] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer chemotherapy is the object of many fundamental and clinical researches. The development in molecular techniques and structural studies at the molecular level have led to the discovery of key proteins involved in the regulation of cell proliferation. This opened perspectives to characterize new anticancer drugs in order to reduce the limitations found with conventional drugs such as the lack of selectivity for cancer cells and resistance phenomena. This review presents the anticancer drugs in clinical investigations that target molecules involved in the signal transduction impairment, the cell cycle deregulation and the differentiation with comments on their mechanisms of action.
Collapse
Affiliation(s)
- E Damiens
- Laboratoire de Chimie Biologique, UMR n(o)111 du CNRS, Villeneuve d'Ascq, France
| |
Collapse
|
70
|
Abstract
Neoplastic cell survival is governed by a balance between pro-apoptotic and anti-apoptotic signals. Noteworthy among several anti-apoptotic signaling elements is the protein kinase C (PKC) isoenzyme family, which mediates a central cytoprotective effect in the regulation of cell survival. Activation of PKC, and subsequent recruitment of numerous downstream elements such as the mitogen-activated protein kinase (MAPK) cascade, opposes initiation of the apoptotic cell death program by diverse cytotoxic stimuli. The understanding that the lethal actions of numerous antineoplastic agents are, in many instances, antagonized by cytoprotective signaling systems has been an important stimulus for the development of novel antineoplastic strategies. In this regard, inhibition of PKC, which has been shown to initiate apoptosis in a variety of malignant cell types, has recently been the focus of intense interest. Furthermore, there is accumulating evidence that selective targeting of PKC may prove useful in improving the therapeutic efficacy of established antineoplastic agents. Such chemosensitizing strategies can involve either (a) direct inhibition of PKC (e.g., following acute treatment with relatively specific inhibitors such as the synthetic sphingoid base analog safingol, or the novel staurosporine derivatives UCN-01 and CGP-41251) or (b) down-regulation (e.g., following chronic treatment with the non-tumor-promoting PKC activator bryostatin 1). In preclinical model systems, suppression of the cytoprotective function(s) of PKC potentiates the activity of cytotoxic agents (e.g., cytarabine) as well as ionizing radiation, and efforts to translate these findings into the clinical arena in humans are currently underway. Although the PKC-driven cytoprotective signaling systems affected by these treatments have not been definitively characterized, interference with PKC activity has been associated with loss of the mitogen-activated protein kinase (MAPK) response. Accordingly, recent pre-clinical studies have demonstrated that pharmacological disruption of the primary MEK-ERK module can mimic the chemopotentiating and radiopotentiating actions of PKC inhibition and/or down-regulation.
Collapse
Affiliation(s)
- W D Jarvis
- Department of Medicine, Medical College of Virginia, Richmond 23298, USA.
| | | |
Collapse
|
71
|
Abstract
Chronic lymphocytic leukaemia (CLL) is the most common form of adult leukaemia in Western countries. The diagnosis requires mature-appearing lymphocytes in the peripheral blood to >5 x 10(9)/L. The immunophenotype typically includes B cell antigens CD19, CD20 and CD23, low expression of surface immunoglobulin and CD5+, with other T cell antigens absent. Bone marrow biopsy, although not required for diagnosis, must show at least 30% lymphocytes. Cytogenetic abnormalities are frequent in patients with CLL, and may be associated with poor prognosis. Clinically, most patients are asymptomatic at presentation, with incidental lymphadenopathy and/or hepatosplenomegaly in the routine physical examination. Infections by opportunistic pathogens are the major cause of death. Aggressive transformation occurs in 10% of patients with CLL, most commonly prolymphocytic leukaemia (PLL) and Richter's syndrome. PLL de novo must be differentiated from PLL of an aggressive transformation. The incidences of autoimmune diseases and solid or haemopoietic secondary malignancies are increased in patients with CLL. Clinical stage is the strongest prognostic factor in CLL. There is no indication for early intervention. The current recommendation to start treatment includes disease-related symptoms, massive and/or progressive hepatosplenomegaly or lymphadenopathy, increasing bone marrow failure, autoimmune disease, and recurrent infections. Alkylating agents (e.g. chlorambucil) and nucleoside analogues (e.g. fludarabine) are the most active agents for CLL. Fludarabine induces higher response rates, but no improvement in overall survival has been observed. Fludarabine is the drug of choice for the majority of patients with CLL. Chlorambucil may be helpful for elderly patients with poor performance, and for patients who do not tolerate fludarabine. No drug combination is better than single agents. For patients refractory to initial treatment, referral to a clinical trial is the best choice. Other salvage therapy includes retreatment with the same initial agent (chlorambucil or fludarabine) if initial response was observed, or fludarabine for patients refractory to chlorambucil. Promising new approaches include cycle-active agents, nelarabine, biological therapy such as anti-CD52 monoclonal antibody, bone marrow transplantation, including the use of submyeloablative preparative regimens ('minitransplant') to induce graft-versus-leukaemia effect, and gene therapy. Prophylactic antibacterials and intravenous immunoglobulin should not be used routinely during supportive care. Epoetin may be helpful for patients who have anaemia without obvious cause. Assessment of response to therapy in CLL has been updated by the National Cancer Institute Working Group, and these guidelines are used worldwide for clinical trials.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- Combined Modality Therapy
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Prolymphocytic/drug therapy
- Leukemia, Prolymphocytic/therapy
Collapse
Affiliation(s)
- N Kalil
- National Cancer Institute, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
72
|
Apoptosis regulating proteins as targets of therapy for haematological malignancies. Expert Opin Investig Drugs 1999; 8:2027-2057. [PMID: 11139839 DOI: 10.1517/13543784.8.12.2027] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Most chemotherapeutic agents used in the treatment of haematological malignancies cause cell death by inducing apoptosis through undefined means. The discovery of the proteins involved in apoptosis and the description of apoptotic pathways suggest new potential targets for therapeutic intervention. Both 'intrinsic' and 'extrinsic' pathways can be activated separately, but activation of caspases appears central to most apoptotic pathways. Novel approaches attempt to induce apoptosis by directly targeting a portion of an apoptotic pathway. Agents that trigger signalling of Fas or tumour necrosis factor- (TNF-) related apoptosis inducing ligand (TRAIL) receptor seek to induce the extrinsic pathway at the cell surface. The BCL-2 family of proteins seems central to the regulation of those apoptotic pathways that involve mitochondrial sequestration or the release of cytochrome c, with subsequent activation of Apaf-1, caspase-9 and caspase-3. The activity of this family may depend upon both the phosphorylation state of different members and the relative level of pro- and anti-apoptotic members. New agents such as the staurosporine analogue UCN-01 and bryostatin are thought to affect apoptosis induction by altering BCL-2 phosphorylation. Others, such as BCL-2 antisense and ATRA attempt to modulate the protein levels to promote apoptosis. Direct activation of caspase-3 is a probable target, but as yet no agent with this direct function is in trial. Clinical trials of several agents have been completed or are underway. It is likely that agents that target particular points in apoptosis pathways will have antileukaemia/lymphoma activity, however, the optimal utilisation may involve combination with other more conventional agents that also activate apoptosis.
Collapse
|
73
|
Pharmacology and clinical experience with bryostatin 1: a novel anticancer drug. Expert Opin Investig Drugs 1999; 8:2189-2199. [PMID: 11139848 DOI: 10.1517/13543784.8.12.2189] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bryostatin 1 (bryo 1) is an example of a novel class of anticancer drug which modulates protein kinase C (PKC) activity. It has varied biological effects mediated largely by the initial activation of PKC, followed by its rapid downregulation. Bryo 1 stimulates in vitro and in vivo haematopoietic progenitor cell growth in a concentration-dependent and lineage-specific fashion. Granulocytes, lymphocytes, monocytes and platelets are all functionally stimulated by bryo 1. Stimulation of cytotoxic T-cell activity by bryo 1 has led to research utilising bryo 1 as an immunotherapeutic agent in mouse tumour xenograft models. The clinical development of bryo 1 followed the demonstration of direct in vitro activity against various tumour cell lines. Multiple Phase I trials have shown muscle pain and flu-like symptoms are the most common toxicities associated with administration of bryo 1. There is particular interest in the role of bryo 1 in haematologic malignancies because of its capacity to induce leukaemic cell differentiation. There is ample in vitro data demonstrating that bryo 1 can sensitise tumour cells to cytotoxic agents. Recent clinical work has focused on combining bryo 1 with traditional chemotherapeutic agents for both haematologic and non-haematologic cancers.
Collapse
|
74
|
Abstract
A large number of cancer chemotherapeutic agents, are in development, many already undergoing clinical testing. A number of these compounds were designed either to modulate or inhibit molecular targets which have been identified as being critical to the development or control of cancer. Targets for inhibition include matrix metalloproteinases, mediators of signal transduction (tyrosine kinases, cyclin dependent kinases and other kinases such as protein kinase C and A) as well as ras expression and prenylation. Classes of potential inhibitory compounds include small molecules, humanized monoclonal antibodies or antisense oligonucleotides. Many of these compounds are relatively well advanced in development. Proof of principle has already been demonstrated in some instances and at least one such compound has been approved for use. Although these new compounds offer exciting opportunities, many bring with them real challenges in terms of the selection of appropriate trial design and surrogate end-points.
Collapse
Affiliation(s)
- L Seymour
- National Cancer Institute of Canada Clinical Trials Group, Queens University, 18 Barrie Street, Kingston, Ontario, K7L 3N6
| |
Collapse
|
75
|
Abstract
The bryostatins are a unique family of cancer chemotherapeutic candidates isolated from marine bryozoa. While their molecular mode of action is not known, these macrolactones exhibit high affinities for protein kinase C (PKC) isozymes, compete for the phorbol ester binding site on PKC, and stimulate kinase activity in vitro and in vivo. Unlike the phorbol esters, they do not act as tumor promoters. Despite promising biological properties, the supply of these compounds is limited by the difficulty of their isolation from natural sources and their synthetic complexity. A new class of bryostatin analogues which retain the putative recognition domain of the bryostatins but are simplified through deletions and modifications in the C1-C14 spacer domain have been designed using computer models. A convergent synthesis has been realized for the production, in gram quantities, of these recognition and spacer domains whose coupling allows for the generation of a range of analogues. The final closure process involves a novel macrotransacetalization reaction which proceeds with complete stereoselectivity. The solution structures of two synthetic analogues were determined by NMR spectroscopy and found to be very similar to the previously reported structures of bryostatins 1 and 10. In addition, these structures appear to indicate that the stereochemistry of the C3 hydroxyl group plays a significant role in the conformation of the macrolactone. All analogues bound strongly to a mixture of PKC isozymes, and several exhibited significant levels of in vitro growth inhibitory activity against human cancer cell lines. Taken together, this work provides important steps toward the development and understanding of simplified, synthetically accessible analogues of the bryostatins as potential chemotherapeutic agents.
Collapse
Affiliation(s)
- P A Wender
- Department of Chemistry, Stanford University, Stanford, CA 94305-5080, USA
| | | | | | | |
Collapse
|
76
|
Evans DA, Carter PH, Carreira EM, Charette AB, Prunet JA, Lautens M. Total Synthesis of Bryostatin 2. J Am Chem Soc 1999. [DOI: 10.1021/ja990860j] [Citation(s) in RCA: 229] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- David A. Evans
- Contribution from the Department of Chemistry & Chemical Biology, Harvard University, Cambridge, Massachusetts 02138
| | - Percy H. Carter
- Contribution from the Department of Chemistry & Chemical Biology, Harvard University, Cambridge, Massachusetts 02138
| | - Erick M. Carreira
- Contribution from the Department of Chemistry & Chemical Biology, Harvard University, Cambridge, Massachusetts 02138
| | - André B. Charette
- Contribution from the Department of Chemistry & Chemical Biology, Harvard University, Cambridge, Massachusetts 02138
| | - Joëlle A. Prunet
- Contribution from the Department of Chemistry & Chemical Biology, Harvard University, Cambridge, Massachusetts 02138
| | - Mark Lautens
- Contribution from the Department of Chemistry & Chemical Biology, Harvard University, Cambridge, Massachusetts 02138
| |
Collapse
|
77
|
Abstract
The ultimate target of pharmacological research is to find new drugs for treating human diseases such as cancer. Agents causing differentiation and thus growth arrest should be particularly useful in this regard. A potential target for such anticancer therapy is the enzyme family protein kinase C (PKC), which is involved in the transduction of signals for cell proliferation, differentiation, and apoptosis. Our recent work showing the induction of differentiation in melanoma cells by an activator of one PKC isoform, PKCdelta, touches on several important areas of investigation, which will form the basis of this review: the role of individual isoforms of PKC, their downstream targets and their specific substrates, the mechanism of activation of specific genes involved in the differentiation process, and the molecular basis for the morphological changes associated with differentiation. The central role that PKC plays in these processes points to the need for a greater understanding of the signalling pathways utilized by individual isoforms of this family of enzymes.
Collapse
Affiliation(s)
- D J Watters
- Queensland Cancer Fund Research Unit, Queensland Institute of Medical Research, P.O. Royal Brisbane Hospital, Herston, Australia.
| | | |
Collapse
|
78
|
Thijsen SF, Schuurhuis GJ, van Oostveen JW, Theijsmeijer AP, van der Hem KG, Odding JH, Dräger AM, Ossenkoppele GJ. Effects of bryostatin-1 on chronic myeloid leukaemia-derived haematopoietic progenitors. Br J Cancer 1999; 79:1406-12. [PMID: 10188883 PMCID: PMC2362710 DOI: 10.1038/sj.bjc.6690225] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Bryostatin-1 belongs to the family of macrocyclic lactones isolated from the marine bryozoan Bugula neritina and is a potent activator of protein kinase C (PKC). Bryostatin has been demonstrated to possess both in vivo and in vitro anti-leukaemic potential. In samples derived from chronic myeloid leukaemia (CML) patients, it has been demonstrated that bryostatin-1 induces a macrophage differentiation, suppresses colony growth in vitro and promotes cytokine secretion from accessory cells. We investigated the effect of bryostatin-1 treatment on colony-forming unit-granulocyte macrophage (CFU-GM) capacity in the presence of accessory cells, using mononuclear cells, as well as in the absence of accessory cells using purified CD34-positive cells. Cells were obtained from 14 CML patients as well as from nine controls. Moreover, CD34-positive cells derived from CML samples and controls were analysed for stem cell frequency and ability using the long-term culture initiating cell (LTCIC) assay at limiting dilution. Individual colonies derived from both the CFU-GM and LTCIC assays were analysed for the presence of the bcr-abl gene with fluorescence in situ hybridization (FISH) to evaluate inhibition of malignant colony growth. The results show that at the CFU-GM level bryostatin-1 treatment resulted in only a 1.4-fold higher reduction of CML colony growth as compared to the control samples, both in the presence and in the absence of accessory cells. However, at the LTCIC level a sixfold higher reduction of CML growth was observed as compared to the control samples. Analysis of the LTCICs at limiting dilution indicates that this purging effect is caused by a decrease in output per malignant LTCIC combined with an increase in the normal stem cell frequency. It is concluded that bryostatin-1 selectively inhibits CML growth at the LTCIC level and should be explored as a purging modality in CML.
Collapse
Affiliation(s)
- S F Thijsen
- Department of Haematology, Free University Hospital, Amsterdam
| | | | | | | | | | | | | | | |
Collapse
|