Anda di halaman 1dari 5

The American Journal of Surgery 186 (2003) 264 –268

Special article

Targeted therapies for the treatment of cancer


Julian A. Kim, M.D.*
Department of General Surgery and Center for Cancer Drug Discovery and Development, Cleveland Clinic Foundation, Desk A80, 9500 Euclid Ave.,
Cleveland, OH 44195, USA

Manuscript received April 30, 2003

Abstract
Background: In contrast to conventional cytotoxic chemotherapy and radiation therapy, a new method of targeted cancer therapeutics is
being directed towards molecular pathways that underlie the malignant phenotype. These therapies target specific tumor cell receptors or
signaling events that are critical to tumor progression while reducing toxicity to normal cells.
Data sources: The purpose of this review is to highlight several examples of novel targeted therapeutics that are currently approved by the
FDA for treatment of patients with cancer. Rituxan is a humanized monoclonal antibody that binds to the CD20 antigen present on B cell
lymphomas and is currently approved for the treatment of patients with relapsed or refractory low-grade CD20 positive follicular lymphoma.
The humanized anti-HER-2/neu herceptin is approved for use in patients with metastatic breast cancer that demonstrates overexpression of
HER-2/neu. Finally, Gleevec is a tyrosine kinase inhibitor that inhibits abl-specific phosphorylation and is approved for use in select patients
with chronic myelogenous leukemia that is refractory to interferon therapy.
Conclusions: The lessons learned from the use of these therapeutics will add to the growing knowledge of mechanistic approaches to the
treatment of patients with cancer based upon targeted therapies, and herald a bright future that will improve the lives of patients with cancer.
© 2003 Excerpta Medica, Inc. All rights reserved.

Keywords: Targeted therapies; Cancer; HER-2 neu; C-kit; Monoclonal antibodies; Rituxan; Herceptin; Gleevec

Conventional cancer treatments such as cytotoxic chemo- tors resulting in cytopenias. These side effects not only
therapy and radiation therapy have been developed based reduce the quality of life of cancer patients, but also limit
upon the observation that malignant cells divide at a more dose intensification and ultimately therapeutic antitumor
rapid rate than the normal cells. For example, ionizing activity.
radiation induces DNA damage that, upon multiple cell The advent of molecular biology techniques that enable
divisions, may lead to errors in transcription and translation rapid isolation and analysis of DNA, RNA, and protein has
resulting in cell death [1]. Similarly, cytotoxic chemother- lead to research efforts that focus on tumor biology at the
apy may interrupt microtubule formation that is essential for cellular and subcellular levels. Identification of pathways
mitotic events that ultimately affect cell survival [2]. It is that are unique to cancer cells has lead to development of
without question that conventional therapies have resulted cancer drug therapeutics that target specific processes es-
in significant survival advantages in patients with breast and sential to tumor cell survival. The purpose of this review is
colon cancer as examples [3–5]. to highlight several examples of novel targeted therapeutics
Despite the benefits derived from the antitumor activity that are currently approved by the FDA for treatment of
of conventional cytotoxic therapies, treatment-related tox- patients with cancer.
icity can substantially reduce quality of life in cancer pa-
tients. Therapies directed against rapidly dividing cells will
result in death of epithelium (such as the lining of the Targeting tumor cell antigens—Rituxan
gastrointestinal tract) or may affect hematopoietic progeni-
Monoclonal antibodies (mAbs) were originally devel-
* Corresponding author. Tel.: ⫹1-216-445-3612; fax: ⫹1-216-445-
oped by Milstein and Kohler in 1975 when they success-
7653. fully fused an antibody-producing B-cell with an immortal-
E-mail address: kimj@ccf.org ized myeloma cell line resulting in a hybridoma [6]. These

0002-9610/03/$ – see front matter © 2003 Excerpta Medica, Inc. All rights reserved.
doi:10.1016/S0002-9610(03)00212-5
J.A. Kim / The American Journal of Surgery 186 (2003) 264 –268 265

Fig. 1. Possible mechanism of action of monoclonal antibodies (Mabs). (A) The antibody has activity by virtue of its specific binding region (Fv) and the
constant region (Fc). (B) Mabs may bind extracellular tumor cell surface receptors resulting in signals that induce caspases and tumor cell apoptosis. (C)
Binding of Mabs to a growth factor receptor may sterically inhibit the binding of the growth factor ligand, resulting in blockade of intracellular signalling.
(D) Recognition of the Fc portion of a Mab bound to a tumor-associated antigen may result in phagocytosis by macrophages that have an Fc receptor. (E)
Finally, the Fc portion of Mab bound to the tumor cell surface may activate the complement cascade, resulting in lysis of the tumor cell.

hybridoma cells could then be cloned and screened for Rituxan mechanism of action
large-scale production of mAb, a discovery that was
awarded the Noble Prize in 1984. Monoclonal antibodies Binding of a mAb to a cell surface receptor can result in
bind specifically to short peptide sequences or antigens with several possible antitumor effects (Fig. 1). The mAb may
remarkable specificity and affinity by virtue of their variable bind a portion of the extracellular domain of that receptor
(Fv) regions. Rapid development of mAbs lead to the avail- that may block ligand-receptor interactions, which in the
ability of reagents that could be used to define cell surface case of growth factor receptors, may result in cell death.
markers that were expressed on subclasses of cells from Alternatively, mAbs may induce cell signaling by virtue of
similar hematopoietic lineage. The resulting cluster desig- binding a specific region of the receptor, with downstream
nation (CD) represented a simple yet robust method of events leading to programmed cell death or apoptosis. Ex-
determining the characteristics or phenotype of cells for perimental evidence suggests that the anti-CD20 mAb Rit-
diagnostic purposes [7]. uxan provide pro-apoptotic signals to B-cell lymphomas as
B-cell non-Hodgkin’s lymphoma (NHL) is highly cur-
one mechanism of its antitumor activity in vivo [8].
able by combined multimodal therapies including chemo-
Binding of anti-CD20 mAbs to the cell surface exposes
therapy, radiation therapy, and in rare cases surgery. How-
the constant region (Fc) of the immunoglobulin, which can
ever, up to 25% of patients either relapsed or were
initiate immunologic mechanisms of antitumor activity. The
refractory to therapy, with many progressing to death. Re-
search of the biology of B-cell lymphomas defined a com- Fc can be recognized and bound by phagocytic cells such as
mon receptor expressed on the cell surface termed CD20. macrophages and engulfed in a process known as antibody-
Despite the fact that CD20 was expressed on normal mature dependent cellular cytotoxicity (ADCC). Alternatively, the
B-cells, efforts were aimed at developing mAbs that would Fc region of the immunoglobulin can also activate the
specifically bind to CD20 as therapeutics for B cell NHL. complement cascade resulting in cell lysis, a process termed
Testing of these anti-CD20 mAbs in vitro and in vivo complement-dependent cytotoxicity (CDC). Recent studies
demonstrated high-affinity binding to B-cell lymphoma have suggested that although B-cell lymphoma cells ap-
cells but not other hematopoietic cells such as T cells. These pear to be uniformly susceptible to ADCC, there appears to
observations lead to the idea that CD20 could be a suitable be differences in sensitivity of the cells to CDC that corre-
target for design of therapies for patients with B-cell lym- lated with clinical antitumor responses after Rituxan therapy
phoma. [9].
266 J.A. Kim / The American Journal of Surgery 186 (2003) 264 –268

Development and approval of Rituxan Herceptin mechanism of action

A major factor limiting repeated dosing of mAbs in There appear to be several possible mechanisms by
humans was that the Fc region of the immunoglobulin which Herceptin exerts its antitumor therapeutic effects
structure was murine in origin. After intravenous adminis- against tumor cells that overexpress HER-2/neu. Since the
tration of these mAbs, patients developed human-antimouse mAb is binding a tumor cell-associated receptor, it may
antibodies (HAMAs) that were shown to complex with render the tumor cell susceptible to ADCC and/or CDC in a
murine mAbs upon repeat administration and in some cases manner similar to Rituxan (Fig. 1). Additionally, herceptin
lead to acute anaphylactic or chronic serum sickness-like may prevent ligation of HER-2/neu with its ligand, a pro-
illness. Molecular techniques allowed for replacement of cess that may induce apoptosis. Finally, there is experimen-
murine Fc with human sequences, thus making them “hu- tal evidence that herceptin may affect the regulation of
manized” and less immunogenic [10]. genes related to angiogenesis such as vascular endothelial
Initial clinical trials using Rituxan demonstrated an ap- growth factor (VEGF) and angiopoietin-1 [18].
proximate 36% objective response rate (complete and par- A critical observation of herceptin activity observed both
tial response) in patients with chemotherapy refractory low- in preclinical experimental animal models and human clin-
grade NHL [11]. Interestingly, although Rituxan therapy did ical trials is the synergistic antitumor effects when used in
result in B-cell lymphopenia due to CD20 expression on combination with cytotoxic chemotherapy [19,20]. Combi-
mature B-cells, this side effect was reversible with discon- nations of herceptin and anthracyclines such as doxorubicin
tinuation of treatment and did not result in significant im- resulted in synergistic antitumor effects against human
munosuppression. A pivotal trial in 166 patients reported an breast cancer xenografts in vivo; similar effects were ob-
objective response rate of 48% (6% complete response and served when used in combination with cisplatin against
42% partial response) with a median duration of response of human ovarian carcinoma xenografts. Herceptin appears to
11.2 months [12]. These data resulted in Food and Drug block DNA repair mechanisms, rendering tumor cells more
Administration approval of Rituxan for the treatment of susceptible to various cytotoxic agents. The effects of com-
patients with relapsed or refractory low-grade CD20 posi- binations of herceptin and anthracyclines may also result in
tive follicular lymphoma. The FDA approval of Rituxan in increased toxicity to normal cells, as evidenced in the hu-
November 1997 represented the first mAb therapy approved man clinical trial results where cardiac toxicity has been
for the treatment of human malignancy, and ushered the observed [21].
modern era of targeted therapeutics for cancer.

Targeting tumor cell growth factor Development and approval of Herceptin


receptors—Herceptin
A humanized anti-HER-2/neu mAb was developed by
Molecular analysis of the pathways that underlie tumor Slamon in collaboration with Genentech and tested in hu-
growth has resulted in defining tumor-associated growth man clinical trials of patients with metastatic breast cancer.
factor receptors as potential targets for therapeutic interven- Several Phase I and II trials demonstrated objective clinical
tion. The epidermal growth factor receptor (EGFR) family and radiographic responses to herceptin therapy, particu-
consists of immunoglobulin-like receptors that, upon bind- larly when used in combination with cytotoxic chemother-
ing with the respective growth factor ligand, result in intra- apy [22,23]. These trials results led to FDA approval of
cellular phosphorylation of transcription factors and in- herceptin for use in combination with paclitaxel in patients
creased DNA synthesis [13]. One of the EGFR family that with metastatic breast cancer in 1998. Herceptin was indi-
was overexpressed on approximately 25% of human breast cated in patients who had breast cancer with overexpression
carcinomas was HER-2/neu, encoded by the gene c-erb B2. of HER-2/neu and had not previously received chemother-
In the late 1980s, Dennis Salmon and William McGuire apy for metastatic disease. The mAb was administered
authored a series of papers published in Science demonstrat- weekly as an intravenous infusion of a loading dose of 4
ing that overexpression of the HER-2/neu proto-oncogene mg/kg and subsequent doses of 2 mg/kg. The results of a
was associated with poor clinical outcome in patients with Phase III trial published in the New England Journal of
breast carcinoma [14,15]. These findings were supported by Medicine in 2001 extended the utility of herceptin in pa-
in vitro analysis of human breast carcinoma cell lines, which tients with metastatic disease [24]. In it, 469 women with
lead to a working hypothesis that HER-2/neu played a metastatic breast cancer that overexpressed HER-2/neu
critical role in the biologic behavior of breast cancer. It has were randomly assigned to receive either standard chemo-
more recently been shown that HER-2/neu overexpression therapy alone or standard chemotherapy in combination
is a predictive marker of response to adjuvant cytotoxic with herceptin. Patients who had not previously received
chemotherapy or hormonal therapy or both, and is also anthracyclines in the adjuvant setting were treated with
predictive of poor prognosis in early stage, node negative either standard chemotherapy consisting of doxorubicin or
patients [16,17]. epirubicin in combination with cyclophosphamide. Those
J.A. Kim / The American Journal of Surgery 186 (2003) 264 –268 267

patients who had been previously treated with anthracy- Gleevec mechanism of action
clines were given paclitaxel as standard chemotherapy. The
addition of herceptin to standard chemotherapy was associ- STI571 was initially tested in vitro and found to have
ated with a significant increase in time to disease progres- TKI activity against abl, PDGF-R, and the c-kit proto-
sion (median 7.4 months versus 4.6 months), and objective oncogene. By virtue of its TKI activity against abl, STI571
response (50% overall response rate versus 32%). Herceptin was effective in inhibiting the proliferation of Ph⫹ cells in
in combination with standard chemotherapy was associated vitro that were derived from a number of patients with
with a lower rate of death at 1 year (22% versus 33%) and CML. Preclinical animal studies demonstrated a dose-de-
increased overall survival (median 25.1 months versus 20.3 pendent reduction in human tumor xenografts that contained
months, P ⫽ 0.01). More recent data suggest that herceptin the bcr-abl gene, again confirming antitumor therapeutic
monotherapy may result in a 26% objective response rate as effects.
a first-line treatment in patients with metastatic breast car-
cinoma that overexpresses HER-2/neu [25]. These encour-
aging results have led to current studies of herceptin as a Development and approval of Gleevec
first-line agent in combination with chemotherapy or hor-
monal therapy, or both, in the adjuvant and neoadjuvant Clinical trials of STI571 were initially performed in
treatment of patients with primary breast cancer. patients with Ph⫹ CML. A phase I study in 83 patients who
had failed conventional interferon therapy were treated with
Targeting tumor cell intracellular signaling daily oral doses and a maximum tolerated dose was not
pathways—Gleevec reached [31]. Amazingly, for patients treated with 300 mg
or more there was a 98% complete response rate, and these
Rational drug design based upon knowledge of intracel- responses were durable for up to 300 days. Phase II clinical
lular signaling pathways that are essential for tumor cell trials confirmed the striking response rate and confirmed the
growth has been the focus of intense study. The advent of lack of serious toxicity, making STI571 arguably the first
rapid and efficient methods of isolation, amplification and small molecule targeted therapeutic for cancer that demon-
analysis of RNA, DNA and protein have allowed for accel- strated specific and potent elimination of tumor cells. Based
erated research into the mechanisms critical to tumor cell upon these data, Gleevec received accelerated FDA ap-
progression and metastasis. proval in May 2001 for the treatment of patients with Ph⫹
Chronic myelogenous leukemia (CML) is a malignancy CML who have failed interferon therapy.
that provided the paradigm by which future efforts of ratio- The remarkable potency and selectivity of STI571 lead
nal drug design will be compared. Cytogenetic studies per- to additional studies in patients with other tumor histolo-
formed decades ago correlated the presence of the Philadel- gies. Gastrointestinal stromal tumors (GIST) are relatively
phia chromosome, an abnormal chromosome developed rare malignancies that have been largely refractory to con-
from a translocation of chromosomes 9 and 22, with devel- ventional cytotoxic chemotherapy regimens. However, ba-
opment of CML [26]. Molecular studies demonstrated that sic studies demonstrated that a significant percentage of
this translocation resulted in the juxtaposition of the genes GIST contained a c-kit proto-oncogene mutation that re-
bcr-abl, which appeared to be central regulators of CML sulted in tyrosine kinase activity. Based upon the observa-
progression into a chronic phase [27]. Subsequent studies tion that STI571 was an inhibitor of c-kit kinase activity,
revealed that bcr-abl resulted in a cytoplasmic fusion pro- early phase clinical trials were initiated in patients with
tein (p210) with significant tyrosine kinase activity, a phe- unresected or metastatic GIST [32]. Again, in the subset of
nomenon that was essentially a switch that was continu- patients whose GIST was associated with a c-kit mutation,
ously “on” in that it was phosphorylating intracellular signal oral STI571 resulted in dramatic tumor regression in a
transduction molecules leading to increased DNA synthesis significant percentage of patients with a tumor histology
and mitogenic processes [28]. that previously had been considered refractory. Gleevec is
Purification of the bcr-abl protein and x-ray crystallog- rapidly being evaluated as a first-line therapy in patients
raphy of the tyrosine kinase domain led to the design of with GIST and other tumors that demonstrate activating
small molecules that could bind and block the phosphory- c-kit mutations.
lation of kinase activity [29]. These small molecules, termed
tyrosine kinase inhibitors or TKIs, are designed to inhibit
tyrosine kinase activity associated with specific receptors or Targeted therapies—potential limitations and future
proto-oncogenes based upon the crystal structure of the targets
kinase domain. The signal transduction inhibitor (STI) 571
of Gleevec was developed by Novartis initially as an inhib- Despite the promising clinical results from the agents
itor of platelet-derived growth factor receptor (PDGF-R) but that have been highlighted, there are still significant limita-
was subsequently discovered to exhibit potent inhibition of tions to the concept of “pathway-specific” targeted thera-
abl-specific tyrosine kinase activity [30]. pies. The first limitation is that these agents are only effec-
268 J.A. Kim / The American Journal of Surgery 186 (2003) 264 –268

tive in tumor types that are dependent upon the pathways [10] Jolliffe LK. Humanized antibodies: enhancing therapeutic utility
that are being inhibited. It is readily apparent that most solid through antibody engineering. Int Rev Immunol 1993;10:241–50.
[11] McLaughlin P, Grillo-Lopez AJ, Link BK, et al. Rituximab chimeric
tumors are the result of numerous genetic mutations, and anti-CD20 monoclonal antibody therapy for relapsed indolent lym-
thus inhibiting a single cellular pathway may not result in phoma: half of patients respond to a four-dose treatment program.
significant therapeutic activity. Design of agents that target J Clin Oncol 1998;16:2825–33.
a number of pathways will possibly increase the therapeutic [12] McLaughlin P, Hagemeister FB, Grillo-Lopez AJ. Rituximab in in-
effect, but also increase the risk of treatment-related toxic- dolent lymphoma: the single-agent pivotal trial. Semin Oncol 1999;
26(suppl 14):79 – 87.
ities. [13] Gullick WJ, Downward J, Parker PJ, et al. The structure and function
The challenges of discovering novel targets for therapeu- of the epidermal growth factor receptor studied by using antisynthetic
tic intervention and designing specific and potent agents peptide antibodies. Proc R Soc Lond B Biol Sci 1985;226:127–34.
remains an area of intense research. It is the early success of [14] Slamon DJ, Godolphin W, Jones LA, et al. Studies of the HER-2/neu
the agents reviewed in this article that have built the foun- proto-oncogene in human breast and ovarian cancer. Science 1989;
244:707–12.
dation for development of newer generations of targeted [15] Slamon DJ, Clark GM, Wong SG, et al. Human breast cancer:
therapies in the future. Intracellular pathways are currently correlation of relapse and survival with amplification of the HER-2/
being targeted by drugs that inhibit ras, c-myc, NF-␬B, and neu oncogene. Science 1987;235:177– 82.
MAPK activity. Growth factor pathways that are currently [16] Press MF, Bernstein L, Thomas PA, et al. HER-2/neu gene amplifi-
being targeted by mAbs and kinase inhibitors include cation characterized by fluorescence in situ hybridization: poor prog-
nosis in node-negative breast carcinomas. J Clin Oncol 1997;15:
EGFR, PDGFR, and vascular endothelial growth factor 2894 –904.
(VEGF) receptor. Many of these agents are in phase I and II [17] Pegram MD, Pauletti G, Slamon DJ. HER-2/neu as a predictive
testing in patients with a variety of malignancies that in- marker of response to breast cancer therapy. Breast Cancer Res Treat
clude breast, lung, pancreatic and colon cancers. The les- 1998;52:65–77.
sons learned from these studies will add to the growing [18] Izumi Y, Xu L, di Tomaso E, et al. Tumour biology: herceptin acts as
an anti-angiogenic cocktail. Nature 2002;416:279 – 80.
knowledge of mechanistic approaches to the treatment of [19] Pietras RJ, Fendly BM, Chazin VR, et al. Antibody to HER-2/neu
patients with cancer based upon targeted therapies, and receptor blocks DNA repair after cisplatin in human breast and
herald a bright future that will improve the lives of patients ovarian cancer cells. Oncogene 1994;9:1829 –38.
with cancer. [20] Pegram M, Hsu S, Lewis G, et al. Inhibitory effects of combinations
of HER-2/neu antibody and chemotherapeutic agents used for treat-
ment of human breast cancers. Oncogene 1999;18:2241–51.
[21] Seidman A, Hudis C, Pierri MK, et al. Cardiac dysfunction in the
References trastuzumab clinical trials experience. J Clin Oncol 2002;20:1215–21.
[22] Baselga J. Clinical trials of Herceptin (trastuzumab). Eur J Cancer
[1] Rydberg B. Radiation-induced DNA damage and chromatin structure. 2001;37(suppl 1):18 –24.
Acta Oncol 2001;40:682–5. [23] Baselga J. Phase I and II clinical trials of trastuzumab. Ann Oncol
[2] Marchetti P, Urien S, Cappellini GA, et al. Weekly administration of 2001;12(suppl 1):49 –51.
paclitaxel: theoretical and clinical basis. Crit Rev Oncol Hematol [24] Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus
2002;44(suppl):3–13. a monoclonal antibody against HER2 for metastatic breast cancer that
[3] Moertel CG. Accomplishments in surgical adjuvant therapy for large overexpresses HER2. N Engl J Med 2001;344:783–92.
bowel cancer. Cancer 1992;70(suppl):1364 –71. [25] Vogel CL, Cobleigh MA, Tripathy D, et al. Efficacy and safety of
[4] Harries M, Gore M. Part I: chemotherapy for epithelial ovarian trastuzumab as a single agent in first-line treatment of HER2-over-
cancer-treatment at first diagnosis. Lancet Oncol 2002;3:529 –36. expressing metastatic breast cancer. J Clin Oncol 2002;20:719 –26.
[5] Fisher B, Redmond C, Legault-Poisson S, et al. Postoperative che- [26] de Klein A, van Kessel AG, Grosveld G, et al. A cellular oncogene is
motherapy and tamoxifen compared with tamoxifen alone in the translocated to the Philadelphia chromosome in chronic myelocytic
treatment of positive-node breast cancer patients aged 50 years and leukaemia. Nature 1982;300:765–7.
older with tumors responsive to tamoxifen: results from the National [27] Strife A, Lambek C, Wisniewski D, et al. Discordant maturation as
Surgical Adjuvant Breast and Bowel Project B-16. J Clin Oncol the primary biological defect in chronic myelogenous leukemia. Can-
1990;8:1005–18. cer Res 1988;48:1035– 41.
[6] Kohler G, Milstein C. Derivation of specific antibody-producing [28] Buckle AM, Mottram R, Pierce A, et al. The effect of Bcr-Abl protein
tissue culture and tumor lines by cell fusion. Eur J Immunol 1976;6: tyrosine kinase on maturation and proliferation of primitive haema-
511–9. topoietic cells. Mol Med 2000;6:892–902.
[7] Lanza F, Moretti S, Latorraca A, et al. Moving towards the definition [29] Buchdunger E, Zimmermann J, Mett H, et al. Inhibition of the Abl
of new clusters of designation at the 6th international workshop on protein-tyrosine kinase in vitro and in vivo by a 2-phenylaminopyri-
human leukocyte differentiation antigens. A brief description of the midine derivative. Cancer Res 1996;56:100 – 4.
recently clustered molecules. Eur J Histochem 1996;40(suppl 1):63– [30] Heinrich MC, Griffith DJ, Druker BJ, et al. Inhibition of c-kit receptor
74. tyrosine kinase activity by STI 571, a selective tyrosine kinase inhib-
[8] Shan D, Ledbetter JA, Press OW. Apoptosis of malignant human B itor. Blood 2000;96:925–32.
cells by ligation of CD20 with monoclonal antibodies. Blood 1998; [31] Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a specific
91:1644 –52. inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leuke-
[9] Pedersen IM, Buhl AM, Klausen P, et al. The chimeric anti-CD20 mia. N Engl J Med 2001;344:1031–7.
antibody rituximab induces apoptosis in B-cell chronic lymphocytic [32] Heinrich MC, Blanke CD, Druker BJ, Corless CL. Inhibition of KIT
leukemia cells through a p38 mitogen activated protein-kinase-depen- tyrosine kinase activity: a novel molecular approach to the treatment
dent mechanism. Blood 2002;99:1314 –9. of KIT-positive malignancies. J Clin Oncol 2002;20:1692–703.

Anda mungkin juga menyukai