551
|
Broxterman HJ, Lankelma J, Hoekman K. Resistance to cytotoxic and anti-angiogenic anticancer agents: similarities and differences. Drug Resist Updat 2003; 6:111-27. [PMID: 12860459 DOI: 10.1016/s1368-7646(03)00026-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Intrinsic resistance to anticancer drugs, or resistance developed during chemotherapy, remains a major obstacle to successful treatment. This is the case both for resistance to cytotoxic agents, directed at malignant cells, and for resistance to anti-angiogenic agents, directed at non-malignant endothelial cells. In this review, we will discuss mechanisms of resistance which have a bearing on both these conceptually different classes of drugs. The complexity of drug resistance, involving drug transporters, such as P-glycoprotein, as well as resistance related to the tissue structure of solid tumors and its consequences for drug delivery is discussed. Possible mechanisms of resistance to endothelial cell-targeted drugs, including inhibitors of the VEGF receptor and EGF receptor family, are reviewed. The resistance of cancer cells as well as endothelial cells related to anti-apoptotic signaling events initiated by cell integrin-matrix interactions is discussed. Current strategies to overcome resistance mechanisms are summarized; they include high-dose chemotherapy, tumor targeting of cytotoxics to improve tumor uptake, low-dose protracted (metronomic) chemotherapy and combinations of classical agents with anti-angiogenic agents. This review discusses primarily literature published in 2001 and 2002.
Collapse
Affiliation(s)
- Henk J Broxterman
- Department of Medical Oncology, VU University Medical Center, BR 232, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | | | | |
Collapse
|
552
|
Dicker AP, Williams TL, Iliakis G, Grant DS. Targeting angiogenic processes by combination low-dose paclitaxel and radiation therapy. Am J Clin Oncol 2003; 26:e45-53. [PMID: 12796615 DOI: 10.1097/01.coc.0000072504.22544.3c] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Tumor growth and angiogenesis are interdependent. Paclitaxel and radiation therapy are commonly used in the clinic, in a number of disease sites, requiring high dosages of both drug and radiation for cure. Paclitaxel (Taxol) is a diterpenoid with antitumor activity against a variety of human neoplasms and can amplify the cytotoxic effect of ionizing radiation in vitro, presumably by inducing arrest at metaphase, known to be a very radiosensitive phase of the cell cycle. Little is known about how angiogenesis is affected by paclitaxel when the combination of paclitaxel and radiation are used. We have evaluated the combination of paclitaxel and radiation at various concentrations, on cytokine-induced angiogenesis in vitro with the goal of determining whether reduction of radiation and paclitaxel doses is possible without sacrificing efficacy. We have found that paclitaxel inhibited endothelial cell proliferation, migration, and tube formation (differentiation) at one-tenth the concentration needed to achieve a similar effect on tumor cell lines. In combination with radiation, inhibition of endothelial cell function was additive and increased twofold. The combination of low-dose paclitaxel and radiation suggests a complementary strategy with potential clinical ramifications to target angiogenesis-dependent malignancies.
Collapse
Affiliation(s)
- Adam P Dicker
- Department of Radiation Oncology, Kimmel Cancer Center, Jefferson Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania 19107-5097, USA.
| | | | | | | |
Collapse
|
553
|
Stoeltzing O, Liu W, Reinmuth N, Fan F, Parry GC, Parikh AA, McCarty MF, Bucana CD, Mazar AP, Ellis LM. Inhibition of integrin alpha5beta1 function with a small peptide (ATN-161) plus continuous 5-FU infusion reduces colorectal liver metastases and improves survival in mice. Int J Cancer 2003; 104:496-503. [PMID: 12584749 DOI: 10.1002/ijc.10958] [Citation(s) in RCA: 166] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Integrin alpha(5)beta(1) is expressed on activated endothelial cells and plays a critical role in tumor angiogenesis. We hypothesized that a novel integrin alpha(5)beta(1) antagonist, ATN-161, would inhibit angiogenesis and growth of liver metastases in a murine model. We further hypothesized that combining ATN-161 with 5-fluorouracil (5-FU) chemotherapy would enhance the antineoplastic effect. Murine colon cancer cells (CT26) were injected into spleens of BALB/c mice to produce liver metastases. Four days thereafter, mice were given either ATN-161 (100 mg/kg, every 3rd day) or saline by intraperitoneal injection, with or without combination of continuous-infusion 5-FU (100 mg/kg/2 weeks), which was started on day 7. On day 20 after tumor cell inoculation, mice were killed and liver weights and number of liver metastases were determined. A follow-up study on survival was also conducted in which mice were randomized to receive ATN-161, 5-FU or ATN-161+5-FU. Combination therapy with ATN-161+5-FU significantly reduced tumor burden (liver weight) and number of liver metastases (p<0.02). Liver tumors in the ATN-161 and ATN-161+5-FU groups had significantly fewer microvessels (p<0.05) than tumors in the control or 5-FU-treated groups. Unlike treatment with either agent alone, ATN-161+5-FU significantly increased tumor cell apoptosis and decreased tumor cell proliferation (p<0.03) and improved overall survival (p<0.03, log-rank test). Targeting integrin alpha(5)beta(1) in combination with 5-FU infusion reduced liver metastases formation and improved survival in this colon cancer model. The enhancement of antineoplastic activity from the combination of anti-angiogenic therapy and chemotherapy may be a promising approach for treating metastatic colorectal cancer.
Collapse
Affiliation(s)
- Oliver Stoeltzing
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston 77030-4009, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
554
|
Abstract
IDN 5390 is a seco-derivative cytostatic taxane. Originally selected for its ability to affect endothelial cell motility, the anti-angiogenic properties of IDN 5390 have been documented in experimental models, in vivo and in vitro. Preclinical studies indicate that, in vivo, oral IDN 5390 has a favorable bioavailability, is well tolerated and shows a significant anti-neoplastic activity on a panel of different tumor models, including paclitaxel-resistant tumors. According to its cytostatic rather than cytotoxic nature, frequent administrations of non-toxic doses have proven to be the optimal schedule for IDN 5390 treatment. Preliminary findings suggest the use of this compound in combination with conventional anti-neoplastic therapy. IDN 5390 can be considered the prototype of a new class of well-tolerated, orally available anti-angiogenic taxane derivatives with cytostatic properties.
Collapse
Affiliation(s)
- Giulia Taraboletti
- Department of Oncology, Mario Negri Institute for Pharmacological Research, Bergamo, Italy.
| | | | | | | |
Collapse
|
555
|
Alaoui-Jamali MA, Qiang H. The interface between ErbB and non-ErbB receptors in tumor invasion: clinical implications and opportunities for target discovery. Drug Resist Updat 2003; 6:95-107. [PMID: 12729807 DOI: 10.1016/s1368-7646(03)00024-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The molecular switches by which malignant cancer cells evolve from a confined to an invasive state are poorly understood, but seem to involve a progressive activation of a signaling network shared by several growth factor receptors and non-receptor molecules. Abnormal expression of ErbB tyrosine kinase receptors, commonly seen in cancer, is an early event in the invasive process, which makes these receptors exciting targets for drug discovery. The past few years have been full of promise for ErbB targeting in the context of receptor overexpression, but also fraught with disappointment as clinical efficacy has often been hampered by potential problems such as the heterogeneity of receptor expression within the same tumor, and the extensive cooperative signaling among ErbB and non-ErbB receptors. Cooperative signaling is a common characteristic of invasive cancer cells, and is believed to dictate the genetic program that controls invasion switches. Molecular studies on the combinatorial signaling involved in tumor invasion are becoming a fertile area for target discovery in cancer. This review discusses how cooperative signaling between ErbB and non-ErbB receptors regulates tumor invasion and hence provides multiple opportunities for drug discovery, and how current therapies and investigational drugs could pave the way to even more potent alternative combinatorial therapeutic approaches for invasive cancers.
Collapse
Affiliation(s)
- Moulay A Alaoui-Jamali
- Department of Medicine, Lady Davis Institute for Medical Research, McGill University, Montreal, Que., Canada.
| | | |
Collapse
|
556
|
Glade-Bender J, Kandel JJ, Yamashiro DJ. VEGF blocking therapy in the treatment of cancer. Expert Opin Biol Ther 2003; 3:263-76. [PMID: 12662141 DOI: 10.1517/14712598.3.2.263] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
It is widely accepted that tumour growth beyond a few cubic millimetres cannot occur without the induction of a new vascular supply. Inhibiting the development of new blood vessels (antiangiogenesis) is a potential approach to cancer therapy that has attracted interest in recent years. In theory, this approach should be relatively selective for tumour cells. The endothelial cells which form new vascular networks in tumours are responding to angiogenic stimuli produced by the tumour, but are themselves genetically normal. Endothelium in normal tissue, by contrast, is usually quiescent. Vascular endothelial growth factor (VEGF) is the best-characterised pro-angiogenic factor. It is virtually ubiquitous in human tumours, and higher levels have been correlated with more aggressive disease. Effective blockade of the VEGF pathway has been demonstrated with multiple agents: neutralising antibody, receptor tyrosine kinase inhibitors, and ribozyme or antisense molecules targeting expression. Promising preclinical data document the potential of these agents for tumour growth inhibition and even tumour regression, yet translation of novel therapeutics targeting the VEGF pathway to the clinic has proved a substantial challenge in itself. While showing clear evidence of antitumour activity over a broad spectrum of experimental tumours, the proper selection, dose, timing and sequence of anti-VEGF treatment in human cancer is not at all obvious. Classic Phase I dose escalation trial design may need to be modified, as higher doses may not be optimal in all patients or for all tumours. In addition, alternate or secondary biological end points (e.g., non-progression) may be needed for early phase studies to document true activity, so as not to abandon effective agents. Recent studies of the neutralising antibody bevacizumab, and small molecule tyrosine kinase inhibitor SU5416, demonstrate that, while unlikely to be effective as monotherapy, incorporation of VEGF blockade into cytotoxic regimens may increase overall response rates. However, incorporation may also produce new toxicities, including thromboembolic complications and bleeding. Newer oral agents, such as SU6668, SU11248, PTK787/ZK222584 and ZD6474, are particularly interesting for their potential for chronic therapy. Future clinical trials are likely to build on past experience with stricter entry criteria, supportive care guidelines and the use of surrogate markers.
Collapse
Affiliation(s)
- Julia Glade-Bender
- Division of Pediatric Surgery, College of Physicians and Surgeons at Columbia University, New York, NY 10032, USA
| | | | | |
Collapse
|
557
|
Abstract
This review assembles the laboratory and clinical evidence that cytotoxic chemotherapy and antiangiogenic therapy are each dependent on endothelial cell apoptosis. During cytotoxic chemotherapy, apoptosis of endothelial cells in the vascular bed of tumors precedes apoptosis of tumor cells, even when the tumor has been made drug resistant. Administration of an angiogenesis inhibitor which is not directly cytotoxic to tumor cells can increase tumor cell apoptosis and inhibit tumor growth by inhibiting endothelial proliferation and migration and/or by inducing endothelial apoptosis. Furthermore, oncogene expression and loss of tumor suppressor gene activity can at once protect tumor cells against apoptosis and increase their angiogenic output. Both of these survival advantages conferred on the tumor can be overcome by antiangiogenic therapy. They can also be overcome by cytotoxic chemotherapy administered on a low dose 'antiangiogenic schedule' which continuously exposes endothelial cells in the tumor bed to the drug. As a result, endothelial apoptosis can be demonstrated to precede tumor cell apoptosis, and tumors regress or are inhibited, whether or not the tumor cells are resistant to the drug, and with little or no host toxicity. In contrast, cytotoxic chemotherapy administered on a 'conventional schedule' of maximal tolerated dose followed by an off-therapy interval, becomes ineffective after drug resistance is acquired. On the basis of these experimental findings, chemotherapy of cancer may possibly be improved-i.e. decreased drug resistance and decreased toxic side-effects-by changing dose and schedule to maximize apoptosis of endothelial cells in the vascular bed of tumors. Further improvement may be achieved by combining angiogenesis inhibitors with 'antiangiogenic chemotherapy'.
Collapse
Affiliation(s)
- Judah Folkman
- Department of Surgery, Children's Hospital and Harvard Medical School, Hunnewell 103 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
558
|
Pratesi G, Laccabue D, Lanzi C, Cassinelli G, Supino R, Zucchetti M, Frapolli R, D'Incalci M, Bombardelli E, Morazzoni P, Riva A, Zunino F. IDN 5390: an oral taxane candidate for protracted treatment schedules. Br J Cancer 2003; 88:965-72. [PMID: 12644838 PMCID: PMC2377083 DOI: 10.1038/sj.bjc.6600784] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2002] [Revised: 11/25/2002] [Accepted: 11/28/2002] [Indexed: 01/24/2023] Open
Abstract
The recognition of the antiangiogenic properties of taxanes provides a basis for novel therapeutic approaches. A prolonged exposure to low drug concentrations has been proposed to be the most suitable approach to exploit the antiangiogenic potential of cytotoxic agents. Such schedule is required to target preferentially slowly dividing endothelial cells. The protracted use of taxanes could benefit from the availability of a taxane endowed with a favourable tolerability profile. Among compounds of a novel series of C-seco taxanes, IDN 5390 was originally selected on the basis of its potent antimotility activity and poor cytotoxicity on endothelial cells. The aim of the study was to investigate the preclinical pharmacologic profile of IDN 5390 in a variety of human tumour xenografts, including ovarian and colon carcinoma and a glioblastoma. IDN 5390, delivered by s.c. injection, daily for 5 days per week, exhibited a high activity against all tumours investigated (tumour growth inhibition was always >85%) in the range of well-tolerated doses. The maximum tolerated dose/injection (MTD), with no signs of systemic or local vesicant toxicity, was 120 mg kg(-1). In contrast, paclitaxel, delivered according to the same schedule, exhibited a variable antitumour efficacy associated with a substantial local toxicity (MTD=10 mg kg(-1)). Considering the remarkable efficacy of IDN 5390 delivered s.c. by protracted treatment schedule, the oral route of administration was further investigated, as the most suitable for daily treatment. Indeed, a good bioavailability of oral IDN 5390 was found. Oral IDN 5390 maintained a substantial efficacy against human tumour xenografts, including paclitaxel-resistant tumours, without loss of potency with respect to s.c. administration. In conclusion, the therapeutic advantages of IDN 5390, over paclitaxel, in protracted daily treatment schedules are represented by the oral efficacy and the high tolerability, which are favourable features to exploit the antiangiogenic potential and to design combinations with other effective agents.
Collapse
Affiliation(s)
- G Pratesi
- Istituto Nazionale Tumori, via Venezian I, Milano, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
559
|
Moehler TM, Ho AD, Goldschmidt H, Barlogie B. Angiogenesis in hematologic malignancies. Crit Rev Oncol Hematol 2003; 45:227-44. [PMID: 12633837 DOI: 10.1016/s1040-8428(02)00135-x] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is defined as the formation of new capillaries from preexisting blood vessels and plays an important role in the progression of solid tumors. Recently a similar relationship has been described in several hematologic malignancies. Expression of the angiogenic peptides vascular endothelial growth factor (VEGF) and basic fibroblast growth factor correlates with clinical characteristics in leukemia and non-Hodgkin's-lymphoma and the serum/plasma concentrations serve as predictors of poor prognosis. Increased bone marrow microvessels in multiple myeloma (MM) are correlated with decreased overall survival. Thalidomide which has antiangiogenic effects and direct cytotoxic effects was found to be effective in MM, myelodysplastic syndrome and acute myeloid leukemia (AML). Preliminary data indicate activity of VEGF-tyrosine kinase inhibitors in AML. Clinical research is now aimed at testing antiangiogenic treatment strategies in several hematologic neoplasms as well as identifying the best candidate patients for specific approaches.
Collapse
Affiliation(s)
- T M Moehler
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | | | | | | |
Collapse
|
560
|
Abstract
Neuroblastoma is a tumour derived from primitive cells of the sympathetic nervous system and is the most common solid tumour in childhood. Interestingly, most infants experience complete regression of their disease with minimal therapy, even with metastatic disease. However, older patients frequently have metastatic disease that grows relentlessly, despite even the most intensive multimodality therapy. Recent advances in understanding the biology and genetics of neuroblastomas have allowed classification into low-, intermediate- and high-risk groups. This allows the most appropriate intensity of therapy to be selected - from observation alone to aggressive, multimodality therapy. Future therapies will focus increasingly on the genes and biological pathways that contribute to malignant transformation or progression.
Collapse
MESH Headings
- Aneuploidy
- Cell Transformation, Neoplastic/genetics
- Child, Preschool
- Chromosomes, Human/genetics
- Chromosomes, Human/ultrastructure
- Forecasting
- Ganglioneuroma/genetics
- Ganglioneuroma/pathology
- Gene Amplification
- Gene Expression Regulation, Neoplastic
- Genes, myc
- Genetic Predisposition to Disease
- Genetic Testing
- Humans
- Infant
- Infant, Newborn
- Loss of Heterozygosity
- Models, Genetic
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Neuroblastoma/classification
- Neuroblastoma/genetics
- Neuroblastoma/pathology
- Neuroblastoma/therapy
- Prognosis
- Receptor, trkA/genetics
- Receptor, trkA/physiology
- Receptor, trkB/genetics
- Receptor, trkB/physiology
- Remission, Spontaneous
- Risk
Collapse
Affiliation(s)
- Garrett M Brodeur
- Division of Oncology, The Children's Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, Pennsylvania 19104-4318, USA.
| |
Collapse
|
561
|
Hahnfeldt P, Folkman J, Hlatky L. Minimizing long-term tumor burden: the logic for metronomic chemotherapeutic dosing and its antiangiogenic basis. J Theor Biol 2003; 220:545-54. [PMID: 12623285 DOI: 10.1006/jtbi.2003.3162] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The general utility of the maximum tolerated dose (MTD) paradigm, a strategy aimed at optimizing the chance of total tumor cell eradication, is here questioned. Evidence to date suggests that for many tumors the potential for eradication is in fact remote, with patients consistently demonstrating tumor cell presence subsequent to MTD treatments having eradicative intent. The failure to eradicate is attributed largely to the heterogeneous nature of the tumor. Heterogeneous cell populations demonstrate short-term refractoriness to up-front dose delivery, but "resensitize" as part of dose recovery, showing increased overall susceptibility to a given series of doses when delivered more evenly spaced. It is demonstrated: (1) that the minimization of total tumor burden, rather than complete eradication, may often be the more practical objective; and (2) that regularly spaced, "metronomic" dosing is the best way to achieve it. As a corollary, it is found that the more efficient ability of the tumor endothelial cells to resensitize following dosing predicts a targeting bias towards the endothelial compartment of a tumor when metronomic dosing is employed. This lends theoretical support to recent empirical studies showing that regularly spaced dosing schedules with no extended rest periods act more antiangiogenically, thereby delaying or avoiding the onset of acquired resistance.
Collapse
Affiliation(s)
- Philip Hahnfeldt
- Dana-Farber Cancer Institute and Department of Radiation Oncology, Harvard Medical School, Boston, MA, 02115, USA.
| | | | | |
Collapse
|
562
|
Keleg S, Büchler P, Ludwig R, Büchler MW, Friess H. Invasion and metastasis in pancreatic cancer. Mol Cancer 2003; 2:14. [PMID: 12605717 PMCID: PMC149416 DOI: 10.1186/1476-4598-2-14] [Citation(s) in RCA: 165] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2002] [Accepted: 01/22/2003] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer remains a challenging disease with an overall cumulative 5-year survival rate below 1%. The process of cancer initiation, progression and metastasis is still not understood well. Invasion and tumor metastasis are closely related and both occur within a tumour-host microecology, where stroma and tumour cells exchange enzymes and cytokines that modify the local extracellular matrix, stimulate cell migration, and promote cell proliferation and tumor cell survival. During the last decade considerable progress has been made in understanding genetic alterations of genes involved in local and systemic tumor growth. The most important changes occur in genes which regulate cell cycle progression, extracellular matrix homeostasis and cell migration. Furthermore, there is growing evidence that epigenetic factors including angiogenesis and lymphangiogenesis may participate in the formation of tumor metastasis. In this review we highlight the most important genetic alterations involved in tumor invasion and metastasis and further outline the role of tumor angiogenesis and lymphangiogenesis in systemic tumor dissemination.
Collapse
Affiliation(s)
- Shereen Keleg
- University of Heidelberg, Department of Surgery, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| | - Peter Büchler
- University of Heidelberg, Department of Surgery, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| | - Roman Ludwig
- University of Heidelberg, Department of Surgery, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| | - Markus W Büchler
- University of Heidelberg, Department of Surgery, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| | - Helmut Friess
- University of Heidelberg, Department of Surgery, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| |
Collapse
|
563
|
Abstract
The use of antiangiogenic therapy is gaining momentum as a novel treatment for a number of conditions, ranging from cancer to psoriasis. This has stemmed from research in the early 1970s showing that the formation of new blood vessels by pre-existing endothelial cells is essential in tumour growth and progression. However, although antiangiogenic therapy was hailed as a new avenue of treatment for cancer, initial clinical data have been disappointing. This has led to the reassessment of antiangiogenic therapy for cancer, and new strategies have been proposed to increase the efficacy of these agents in this setting. Angiogenesis has also been implicated in other conditions that are notoriously difficult to treat, such as arteriosclerosis, arthritis, psoriasis and diabetic retinopathy. Increased understanding of the angiogenic process, the diversity of its inducers and mediators, appropriate drug schedules and the use of these agents with other modalities may lead to radically new treatment regimens for many of these conditions. The role of angiogenesis in different pathological settings, and emerging antiangiogenic agents currently in preclinical and clinical studies are discussed in this review. However, while potential benefits are profound, limitations of antiangiogenic therapy have also been identified, suggesting that there is also a need for caution in applying these compounds to the clinical setting.
Collapse
Affiliation(s)
- Keith Dredge
- Division of Oncology, St. George's Hospital Medical School, Jenner Wing, Cranmer Terrace, Tooting SW17 0RE, London, UK.
| | | | | |
Collapse
|
564
|
Abstract
Angiogenesis inhibitors are a new class of drugs, for which the general rules involving conventional chemotherapy might not apply. The successful translation of angiogenesis inhibitors to clinical application depends partly on the transfer of expertise from scientists who are familiar with the biology of angiogenesis to clinicians. What are the most common questions that clinicians ask as they begin to test angiogenesis inhibitors in cancer clinical trials?
Collapse
Affiliation(s)
- Robert Kerbel
- Molecular and Cellular Biology Research, Sunnybrook and Women's College Health Sciences Centre, S-218, 2075 Bayview Avenue, Toronto, Ontario M4N 3M5, Canada.
| | | |
Collapse
|
565
|
Abstract
Targeting angiogenesis represents a new strategy for the development of anticancer therapies. New targets derived from proliferating endothelial cells may be useful in developing anticancer drugs that prolong or stabilize the progression of tumors with minimal systemic toxicities. These drugs may also be used as novel imaging and radiommunotherapeutic agents in cancer therapy. In this review, the mechanisms and control of angiogenesis are discussed. Genetic and proteomic approaches to defining new potential targets on tumor vasculature are then summarized, followed by discussion of possible antiangiogenic treatments that may be derived from these targets and current clinical trials. Such strategies involve the use of endogenous antiangiogenic agents, chemotherapy, gene therapy, antiangiogenic radioligands, immunotherapy, and endothelial cell-based therapies. The potential biologic end points, toxicities, and resistance mechanisms to antiangiogenic agents must be considered as these therapies enter clinical trials.
Collapse
Affiliation(s)
- Frank A Scappaticci
- Department of Pathology, Stanford University Medical Center, Stanford, CA 94305, USA.
| |
Collapse
|
566
|
Presta M, Belleri M, Vacca A, Ribatti D. Anti-angiogenic activity of the purine analog 6-thioguanine. Leukemia 2002; 16:1490-9. [PMID: 12145690 DOI: 10.1038/sj.leu.2402646] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2002] [Accepted: 05/14/2002] [Indexed: 11/09/2022]
Abstract
The antimetabolite 6-thioguanine (6-TG) is utilized in the management of acute myelogenous leukemia (AML). Angiogenesis is a possible therapeutic target in hematologic tumors. Thus, we addressed the possibility that 6-TG may also act as an anti-angiogenic molecule. 6-TG inhibited endothelial cell proliferation triggered by fibroblast growth factor-2 (FGF2) and vascular endothelial growth factor (VEGF) and delayed the repair of a mechanically wounded endothelial cell monolayer. Also, 6-TG inhibited sprouting within fibrin gel, morphogenesis on Matrigel, and collagen gel invasion by endothelial cells. 2-Aminopurine was ineffective. In vivo, 6-TG inhibited basal, VEGF-induced, and FGF2-induced vascularization in the chick embryo chorioallantoic membrane and prevented neovascularization triggered by leukemia LIK cells or their conditioned medium. Finally, bone marrow vascularization in AML patients was decreased to control values in the early remission phase and persisted unvaried after 8-12 months of maintenance therapy with 6-TG. Thus, 6-TG inhibits different steps of the angiogenesis process in vitro and exerts a potent anti-angiogenic activity in vivo. Its anti-angiogenic activity, together with its antimetabolite activity towards tumor cells, may contribute to its action during maintenance therapy in AML. These results suggest a new rationale for the use of purine analogs in the management of AML.
Collapse
Affiliation(s)
- M Presta
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, Italy
| | | | | | | |
Collapse
|
567
|
Tran J, Master Z, Yu JL, Rak J, Dumont DJ, Kerbel RS. A role for survivin in chemoresistance of endothelial cells mediated by VEGF. Proc Natl Acad Sci U S A 2002; 99:4349-54. [PMID: 11917134 PMCID: PMC123651 DOI: 10.1073/pnas.072586399] [Citation(s) in RCA: 316] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2001] [Indexed: 01/13/2023] Open
Abstract
Although standard anticancer chemotherapeutic drugs have been designed to inhibit the survival or growth of rapidly dividing tumor cells, it is possible to enhance the efficacy of such drugs by targeting the proliferating host endothelial cells (ECs) of the tumor vasculature. A theoretical advantage of this strategy lies in the possibility of circumventing, or significantly delaying, acquired drug resistance driven by the genetic instability of tumor cells. Here, we show that both vascular endothelial growth factor (VEGF) and basic fibroblast growth factor significantly reduce the pro-apoptotic potency of chemotherapy on both micro- and macrovascular ECs. This cytoprotection to drug toxicity was found to be phosphatidylinositol 3-kinase-dependent and could be recapitulated in the absence of VEGF by overexpressing the dominant-active form of the serine/threonine kinase protein kinase B/Akt. Downstream of phosphatidylinositol 3-kinase, we also show that survivin plays a pivotal role in VEGF-mediated EC protection by preserving the microtubule network. In this respect, its induction effectively protects ECs against chemotherapeutic damage, whereas overexpression of its dominant-interfering mutant (C84A) abrogates the protective effects of VEGF. Accordingly, the potency of VEGF as a chemoprotectant was more pronounced with drugs that interfere with microtubule dynamics than those that damage DNA. These studies implicate a role for survivin up-regulation as a novel mechanism of EC drug "resistance" and support the notion that angiogenic factors that induce the expression of survivin may act to shield tumor ECs from the apoptotic effects of chemotherapy. Thus, exploiting chemotherapeutic drugs as antiangiogenics is likely to be compromised by the high concentrations of proangiogenic survival/growth factors present in the tumor microenvironment; targeting EC survival pathways should improve the antiangiogenic efficacy of antineoplastic agents, particularly microtubule-inhibitor drugs.
Collapse
Affiliation(s)
- Jennifer Tran
- Molecular and Cellular Biology Research, Sunnybrook and Women's College Health Sciences Centre, Toronto, ON, Canada M4N 3M5
| | | | | | | | | | | |
Collapse
|
568
|
|
569
|
Kerbel RS, Klement G, Pritchard KI, Kamen B. Continuous low-dose anti-angiogenic/ metronomic chemotherapy: from the research laboratory into the oncology clinic. Ann Oncol 2002; 13:12-5. [PMID: 11863092 DOI: 10.1093/annonc/mdf093] [Citation(s) in RCA: 133] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
570
|
Abstract
As a model system for the understanding of human cancer, the mouse has proved immensely valuable. Indeed, studies of mouse models have helped to define the nature of cancer as a genetic disease and demonstrated the causal role of genetic events found in tumors. As the scientific and medical community's understanding of human cancer becomes more sophisticated, however, limitations and potential weaknesses of existing models are revealed. How valid are these murine models for the understanding and treatment of human cancer? The answer, it appears, depends on the nature of the research requirement. Certain models are better suited for particular applications. Using novel molecular tools and genetic strategies, improved models have recently been described that accurately mimic many aspects of human cancer.
Collapse
Affiliation(s)
- B Hann
- Cancer Research Institute, Mount Zion Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA 94143-0875, USA
| | | |
Collapse
|
571
|
Abstract
Tumour endothelium is a new target for anticancer treatments. Proliferating endothelial cells from the tumour, even if qualitatively different from those of blood vessels in the normal tissue of origin, remain putatively normal and genetically stable cells. The results of recent experimental studies have suggested that frequent administration of certain cytotoxic agents at low doses (a tenth to a third of the maximum tolerated dose), known as 'metronomic' chemotherapy, increases the antiangiogenic activity of the drugs. The effects of these metronomic schedules of cytotoxic agents may be further enhanced by concurrent administration of novel, selective, treatments that inhibit, at a molecular level, the processes of tumour formation and growth eg angiogenesis, growth factor pathways, and other signal transduction cascades. The need to treat patients for long periods also supports the use of metronomic scheduling for chemotherapy, to minimise toxicity and to target both proliferating tumour cells and endothelial cells. This review describes the experimental studies involving metronomic schedules of chemotherapy, alone and in combination with angiogenesis inhibitors, and suggests a new therapeutic anticancer paradigm for controlling cancer by long-term therapy, based on the development of combinations of metronomic cytotoxic agents with individually tailored compounds designed to target specific molecules.
Collapse
Affiliation(s)
- G Gasparini
- Division of Medical Oncology, Azienda Complesso Ospedaliero San Filippo Neri, Rome, Italy.
| |
Collapse
|
572
|
Kruczynski A, Hill BT. Vinflunine, the latest Vinca alkaloid in clinical development. A review of its preclinical anticancer properties. Crit Rev Oncol Hematol 2001; 40:159-73. [PMID: 11682323 DOI: 10.1016/s1040-8428(01)00183-4] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Vinflunine is a new Vinca alkaloid uniquely fluorinated, by the use of superacid chemistry, in a little exploited region of the catharanthine moiety. In vitro investigations have confirmed the mitotic-arresting and tubulin-interacting properties of vinflunine shared by other Vinca alkaloids. However, differences in terms of the inhibitory effects of vinflunine on microtubules dynamics and its tubulin binding affinities have been identified which appear to distinguish it from the other Vinca alkaloids. Vinflunine induced smaller spirals with a shorter relaxation time, effects, which might be associated with reduced neurotoxicity. Studies investigating the in vitro cytotoxicity of vinflunine in combination therapy have revealed a high level of synergy when vinflunine was combined with either cisplatin, mitomycin C, doxorubicin or 5-fluorouracil. Furthermore, although vinflunine appears to participate in P-glycoprotein-mediated drug resistance mechanisms, it has proved only a weak substrate for this protein and a far less potent inducer of resistance than vinorelbine. Vinflunine was identified in preclinical studies as having marked antitumour activity in vivo against a large panel of experimental tumour models, with tumour regressions being recorded in human renal and small cell lung cancer tumour xenografts. Overall its level of activity was superior to that of vinorelbine in many of the experimental models used. Interestingly, an in vivo study using a well vascularised adenocarcinoma of the colon has suggested that vinflunine mediates its antitumour activity at least in part via an antivascular mechanism, even at sub-cytotoxic doses. Therefore, these data provide a favourable preclinical profile for vinflunine, supporting its promising candidacy for clinical development. Phase I evaluations of vinflunine have been completed in Europe and phase II clinical trials are now ongoing.
Collapse
MESH Headings
- Adenocarcinoma/blood supply
- Angiogenesis Inhibitors/pharmacology
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Antineoplastic Agents, Phytogenic/administration & dosage
- Antineoplastic Agents, Phytogenic/adverse effects
- Antineoplastic Agents, Phytogenic/chemistry
- Antineoplastic Agents, Phytogenic/pharmacokinetics
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Apoptosis/drug effects
- Cell Line/drug effects
- Colonic Neoplasms/blood supply
- DNA Damage
- Drug Resistance, Neoplasm
- Drug Screening Assays, Antitumor
- Drug Synergism
- Forecasting
- Humans
- Leukemia P388/drug therapy
- Lung Neoplasms/drug therapy
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/secondary
- Mice
- Microtubules/drug effects
- Microtubules/ultrastructure
- Molecular Structure
- Neoplasms, Experimental/drug therapy
- Nervous System Diseases/chemically induced
- Tubulin Modulators
- Tumor Cells, Cultured/drug effects
- Vinblastine/administration & dosage
- Vinblastine/adverse effects
- Vinblastine/analogs & derivatives
- Vinblastine/chemistry
- Vinblastine/pharmacokinetics
- Vinblastine/pharmacology
- Vinblastine/therapeutic use
- Vinca Alkaloids/pharmacology
- Vinca Alkaloids/therapeutic use
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- A Kruczynski
- Division de Cancérologie Expérimentale, Centre de Recherche Pierre Fabre, 17 avenue Jean Moulin, 81106 Cedex 06, Castres, France.
| | | |
Collapse
|
573
|
Pui CH, Campana D, Evans WE. Childhood acute lymphoblastic leukaemia--current status and future perspectives. Lancet Oncol 2001; 2:597-607. [PMID: 11902549 DOI: 10.1016/s1470-2045(01)00516-2] [Citation(s) in RCA: 206] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The current cure rate of 80% in childhood acute lymphoblastic leukaemia attests to the effectiveness of risk-directed therapy developed through well-designed clinical trials. In the past decade there have been remarkable advances in the definition of the molecular abnormalities involved in leukaemogenesis and drug resistance. These advances have led to the development of promising new therapeutic strategies, including agents targeted to the molecular lesions that cause leukaemia. The importance of host pharmacogenetics has also been recognised. Thus, genetic polymorphisms of certain enzymes have been linked with host susceptibility to the development of de novo leukaemia or therapy-related second cancers. Furthermore, recognition of inherited differences in the metabolism of antileukaemic agents has provided rational selection criteria for optimal drug dosages and scheduling. Treatment response assessed by measurements of submicroscopic leukaemia (minimal residual disease) has emerged as a powerful and independent prognostic indicator for gauging the intensity of therapy. Ultimately, treatment based on biological features of leukaemic cells, host genetics, and the amount of residual disease should improve cure rates further.
Collapse
Affiliation(s)
- C H Pui
- Leukaemia/Lymphoma Division, Fahad Nassar Al-Rashid Chair of Leukaemia Research at St Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | | | | |
Collapse
|
574
|
Quirici N, Soligo D, Caneva L, Servida F, Bossolasco P, Deliliers GL. Differentiation and expansion of endothelial cells from human bone marrow CD133(+) cells. Br J Haematol 2001; 115:186-94. [PMID: 11722432 DOI: 10.1046/j.1365-2141.2001.03077.x] [Citation(s) in RCA: 268] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We report a method of purifying, characterizing and expanding endothelial cells (ECs) derived from CD133(+) bone marrow cells, a subset of CD34(+) haematopoietic progenitors. Isolated using immunomagnetic sorting (mean purity 90 +/- 5%), the CD133(+) bone marrow cells were grown on fibronectin-coated flasks in M199 medium supplemented with fetal bovine serum (FBS), vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF) and insulin growth factor (IGF-1). The CD133(+) fraction contained 95 +/- 4% CD34(+) cells, 3 +/- 2% cells expressing VEGF receptor (VEGFR-2/KDR), but did not express von Willebrand factor (VWF), VE-cadherin, P1H12 or TE-7. After 3 weeks of culture, the cells formed a monolayer with a typical EC morphology and expanded 11 +/- 5 times. The cells were further purified using Ulex europaeus agglutinin-1 (UEA-1)-fluorescein isothiocyanate (FITC) and anti-FITC microbeads, and expanded with VEGF for a further 3 weeks. All of the cells were CD45(-) and CD14(-), and expressed several endothelial markers (UEA-1, VWF, P1H12, CD105, E-selectin, VCAM-1 and VE-cadherin) and typical Weibel-Palade bodies. They had a high proliferative potential (up to a 2400-fold increase in cell number after 3 weeks of culture) and the capacity to modulate cell surface antigens upon stimulation with inflammatory cytokines. Purified ECs were also co-cultivated with CD34(+) cells, in parallel with a purified fibroblastic cell monolayer. CD34(+) cells (10 x 10(5)) gave rise to 17,951 +/- 2422 CFU-GM colonies when grown on endothelial cells, and to 12,928 +/- 4415 CFU-GM colonies on fibroblast monolayers. The ECs also supported erythroid blast-forming unit (BFU-E) colonies better. These results suggest that bone marrow CD133(+) progenitor cells can give rise to highly purified ECs, which have a high proliferative capacity, can be activated by inflammatory cytokines and are superior to fibroblasts in supporting haematopoiesis. Our data support the hypothesis that endothelial cell progenitors are present in adult bone marrow and may contribute to neo-angiogenesis.
Collapse
Affiliation(s)
- N Quirici
- Fondazione Matarelli, Ospedale Fatebenefratelli e Oftalmico, Milan, Italy
| | | | | | | | | | | |
Collapse
|
575
|
Tunici P, Gianni D, Finocchiaro G. Gene therapy of glioblastomas: from suicide to homicide. PROGRESS IN BRAIN RESEARCH 2001; 132:711-9. [PMID: 11545030 DOI: 10.1016/s0079-6123(01)32112-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Affiliation(s)
- P Tunici
- Unit of Neuro-Oncology and Gene Therapy, Istituto Nazionale Neurologico Besta, Via Celoria 11, 20133 Milan, Italy
| | | | | |
Collapse
|
576
|
Soffer SZ, Moore JT, Kim E, Huang J, Yokoi A, Manley C, O'Toole K, Stolar C, Middlesworth W, Yamashiro DJ, Kandel JJ. Combination antiangiogenic therapy: increased efficacy in a murine model of Wilms tumor. J Pediatr Surg 2001; 36:1177-81. [PMID: 11479851 DOI: 10.1053/jpsu.2001.25747] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND/PURPOSE Antibody to vascular endothelial growth factor (anti-VEGF) suppresses tumor growth and metastasis in experimental Wilms tumor. However, tumor growth accelerates if antibody is withdrawn. As recently shown, low-dose, frequently administered topotecan, a topoisomerase-1 inhibitor, has anti-angiogenic activity. The authors hypothesized that combined topotecan/anti-VEGF therapy would suppress tumor growth and metastasis more durably than either agent alone. METHODS Xenografts were induced by intrarenal injection of human Wilms tumor cells in athymic mice (n = 59). Mice were divided into control (n = 10), anti-VEGF (n = 16), topotecan (n = 17), and topotecan plus anti-VEGF (n = 16) groups. All control and half the treated mice were killed at week 6. Remaining ("rebound") mice were maintained without treatment until week 8. Tumor vasculature was mapped by fluorescein angiography/PECAM immunostaining. Endothelial apoptosis was assessed by TUNEL assay. RESULTS 6 weeks: Tumor weights were reduced significantly in treated mice (P <.003 v control). Seven of ten control and 1 of 25 treated mice displayed lung metastases (P <.003). Rebound tumors were largest in topotecan-only, intermediate in antibody-treated, and smallest in combination-treated mice. Immunostaining and angiography results showed sparse vascularity in treated xenografts. Endothelial apoptosis was observed only in treated tumors. CONCLUSION Combination low-dose topotecan and anti-VEGF antibody therapy is antiangiogenic and suppresses tumor growth and metastasis in experimental Wilms tumor more durably than either agent alone.
Collapse
Affiliation(s)
- S Z Soffer
- Division of Pediatric Surgery, and the Department of Pathology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
577
|
Burke PA, DeNardo SJ. Antiangiogenic agents and their promising potential in combined therapy. Crit Rev Oncol Hematol 2001; 39:155-71. [PMID: 11418313 DOI: 10.1016/s1040-8428(01)00115-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
One of the most promising strategies for treating cancer is the addition of antiangiogenic therapy to therapeutic regimens. Angiogenesis, or the growth of new blood vessels from preexisting vessels, is essential both for the growth of a primary tumor and for successful metastasis. As a result of intense research in this field, a number of antiangiogenic agents have been identified and have demonstrated varying degrees of success in inhibiting the growth of solid tumors and metastases in preclinical and clinical studies. The real potential of antiangiogenic agents for cancer therapy resides in strategic combinations with each other, with chemotherapy, with radiation, and with tumor-targeting agents, such as radioimmunotherapy. Along with this new opportunity to develop synergistic therapy comes the challenging complexities of the physiologic systems regulating angiogenesis. These multifaceted systems could intimidate investigators seeking to take advantage of the potential synergy in combined cancer therapy. To aid in these efforts, this overview of key antiangiogenic agent mechanisms, combination strategies and initial studies of the potential synergy with chemotherapy, radiation and radioimmunotherapy is presented.
Collapse
Affiliation(s)
- P A Burke
- Division of Hematology/Oncology, Department of Internal Medicine, Davis Medical Center, University of California, 1508 Alhambra Boulevard, Sacramento, CA 95816, USA
| | | |
Collapse
|
578
|
|
579
|
Abstract
Vascular morphogenesis through mechanisms of vasculogenesis, angiogenesis and intussusception is associated primarily with embryonic and fetal development and is down-regulated in the healthy adult. Physiological angiogenesis in the adult is restricted to the female reproductive system where it occurs cyclically in the ovary and the uterus as well as pregnancy-associated in the placenta and in the mammary gland. Of all the different organs, the cyclic corpus luteum of the ovary is the organ site with the strongest physiological angiogenesis. The hormonally regulated cyclic processes in the corpus luteum are characterized by discrete phases of blood vessel growth, vessel maturation and vessel regression. This chapter discusses the morphological changes of the vasculature in the cyclic corpus luteum in relation to the regulating molecular mechanisms. These data establish the dynamic processes in the ovarian corpus luteum as a unique system for studying all steps of the angiogenic cascade, including vessel maturation and vessel regression. Inhibition of angiogenesis impairs the normal ovarian cycle, reflecting that angiogenesis is rate-limiting for ovulation and growth of the corpus luteum and may, thus, be a potential target for therapeutic intervention in the reproductive function.
Collapse
Affiliation(s)
- H G Augustin
- Cell Biology Laboratory, Department of Obstetrics and Gynaecology, University of Göttingen Medical School, Göttingen, D-37075, Germany
| |
Collapse
|
580
|
Kamen BA, Rubin E, Aisner J, Glatstein E. High-time chemotherapy or high time for low dose. J Clin Oncol 2000; 18:2935-7. [PMID: 10944125 DOI: 10.1200/jco.2000.18.16.2935] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|