Orthotopic transplant mouse models with green fluorescent protein-expressing cancer cells to visualize metastasis and angiogenesis

Similar documents
Multi-organ metastatic capability of Chinese hamster ovary cells revealed by green fluorescent protein (GFP) expression

Anumber of approaches have been taken to label

MELANOMA CANCER TEST

Antimetastatic efficacy of adjuvant gemcitabine in a pancreatic cancer orthotopic model

HISASHI ISHIKURA, KAZUYA KONDO, TAKANORI MIYOSHI, YUJI TAKAHASHI, HARUHIKO FUJINO and YASUMASA MONDEN

Chronologically-specific metastatic targeting of human pancreatic tumors in orthotopic models

Genetically Fluorescent Melanoma Bone and Organ Metastasis Models 1

Determination of Clonality of Metastasis by Cell-Specific Color-Coded Fluorescent-Protein Imaging

200 W. Arbor Dr., San Diego, CA , USA. Key words: angiogenesis, metastasis, orthotopic fragment transplant, renal cancer

Current methods of external imaging of internally growing. Whole-body optical imaging of green fluorescent protein-expressing tumors and metastases

In vivo Color-Coded Imaging of the Interaction of Colon Cancer Cells and Splenocytes in the Formation of Liver Metastases

INTRODUCTION. Journal of Surgical Research 132, (2006) doi: /j.jss

this mutation. However, VEM was not effective. The PDOX model thus helped identify

Whole-Body Subcellular Multicolor Imaging of Tumor-Host Interaction and Drug Response in Real Time

Hair Follicle Derived Blood Vessels Vascularize Tumors in Skin and Are Inhibited by Doxorubicin

DAWNING OF THE AGE OF ANGIOGENESIS

ANTICANCER RESEARCH 32: (2012)

Supplementary Materials. for Garmy-Susini, et al, Integrin 4 1 signaling is required for lymphangiogenesis and tumor metastasis

Tumor-educated Macrophages Promote Tumor Growth and Peritoneal Metastasis in an Orthotopic Nude Mouse Model of Human Pancreatic Cancer

Juyoun Jin, D.V.M., Ph.D. Institute for Refractory Cancer Research, Samsung Medical Center

Color-Coded Fluorescent Protein Imaging of Angiogenesis: The AngioMouse Models

Nestin-Linked Green Fluorescent Protein Transgenic Nude Mouse for Imaging Human Tumor Angiogenesis

Species Tumor Type Comment for in vivo work Lead Time for in vivo studies [weeks] MB-49-luc-2 Mouse urinary bladder carcinoma C57BL/6 2

Optimization of the Fuse-It-mRNA Protocol for L929 Cells in the µ-plate 24 Well

ANTICANCER RESEARCH 35: (2015)

(A) RT-PCR for components of the Shh/Gli pathway in normal fetus cell (MRC-5) and a

Dual-color Imaging of Nascent Angiogenesis and its Inhibition in Liver Metastases of Pancreatic Cancer

Cellular expression of green fluorescent protein, coupled with high-resolution in vivo videomicroscopy, to monitor steps in tumor metastasis

1ME A17R11 WI238 DMS2114 JC L2929 P815 PT67

B16-F10 (Mus musculus skin melanoma), NCI-H460 (human non-small cell lung cancer

Supplemental Table S1

Supplemental Material

(a) Schematic diagram of the FS mutation of UVRAG in exon 8 containing the highly instable

A Nod Scid mouse model to study human prostate cancer

TITLE: Anti-Angiogenic Gene Therapy of Prostate Cancer with Lentiviral Vectors

Interactive Staging Bee

Efficacy of Dietary Antioxidants Combined with a Chemotherapeutic Agent on Human Colon Cancer Progression in a Fluorescent Orthotopic Mouse Model

Establishment of a malignant pleural effusion mouse model with lewis lung carcinoma cell lines expressing enhanced green fluorescent protein

Construction of a hepatocellular carcinoma cell line that stably expresses stathmin with a Ser25 phosphorylation site mutation

Argininosuccinate synthetase 1 suppression and arginine restriction inhibit cell

Sample validation data

Can we prevent metastasis?

(a) Significant biological processes (upper panel) and disease biomarkers (lower panel)

Cancer Biology Course. Invasion and Metastasis

VEGF receptor antisense therapy inhibits angiogenesis and peritoneal dissemination of human gastric cancer in nude mice

Figure S1. Generation of inducible PTEN deficient mice and the BMMCs (A) B6.129 Pten loxp/loxp mice were mated with B6.

Definition of Synoptic Reporting

The Cyan Fluorescent Protein Nude Mouse as a Host for Multicolor-coded Imaging Models of Primary and Metastatic Tumor Microenvironments

Development of the Transgenic Cyan Fluorescent Protein (CFP)-Expressing Nude Mouse for Technicolor Cancer Imaging

Molecular Imaging for Cancer Visualization

Imaging Nuclear Cytoplasmic Dynamics in Primary and Metastatic Colon Cancer in Nude Mice

MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells

Surgery remains the mainstay treatment for localized

Neoplasia 2018 Lecture 2. Dr Heyam Awad MD, FRCPath

Establishment of in Vivo Metastasis Model of Human Adenoid Cystic Carcinoma: Detection of Metastasis by PCR with Human β -Globin Gene

A549 and A549-fLuc cells were maintained in high glucose Dulbecco modified

Interactions between cancer stem cells and their niche govern metastatic colonization

Neoplasia part I. Dr. Mohsen Dashti. Clinical Medicine & Pathology nd Lecture

Integrin v 3 targeted therapy for Kaposi s sarcoma with an in vitro evolved antibody 1

An Imageable Metastatic Treatment Model of Nasopharyngeal Carcinoma

Invasion And Metastasis. Wirsma Arif Harahap Surgical Oncologist Surgery Department Andalas Medical Schoool

Supplementary Appendix

(A) Cells grown in monolayer were fixed and stained for surfactant protein-c (SPC,

In vivo imaging with fluorescent proteins: the new cell biology $

Supplementary Fig. 1: ATM is phosphorylated in HER2 breast cancer cell lines. (A) ATM is phosphorylated in SKBR3 cells depending on ATM and HER2

Bronchogenic Carcinoma

ANTICANCER RESEARCH 36: (2016)

Recruitment of Cancer-Associated Fibroblasts and Blood Vessels by Orthotopic Liver Tumors Imaged in Red Fluorescent Protein (RFP) Transgenic Nude Mice

Beverly A. Teicher, PhD DCTD/NCI. The content reflects my professional opinions, not an NCI policy statement.

Electron micrograph of phosphotungstanic acid-stained exosomes derived from murine

Supplementary Table 1. Characterization of HNSCC PDX models established at MSKCC

ITO, Yasuhiro ; FUJII, Mizue ; SHIBUYA, Takashi ; UEHARA, Jiro ; SATO, Katsuhiko ; IIZUKA, Hajime

Cancer Metronomic Therapy Milan, February 26, 2016

Tumor DNA circulating in the plasma might play a role in metastasis. The hypothesis of the genometastasis

An epithelial-to-mesenchymal transition-inducing potential of. granulocyte macrophage colony-stimulating factor in colon. cancer

AJCC-NCRA Education Needs Assessment Results

For personal use only

Real-time imaging reveals the single steps of brain metastasis fo mation r

Inhibitors of Methionine Aminopeptidase-2 2 in the Treatment of Non-Hodgkin

Date Lab Pd. Lecture Notes (57)

TITLE: Systemic Oncolytic Cytokine HSV Therapy of Prostate Cancer. CONTRACTING ORGANIZATION: Massachusetts General Hospital Boston, MA

Supporting information. Precise Photodynamic Therapy of Cancer via Subcellular Dynamic Tracing of Dual-loaded Upconversion Nanophotosensitizers

SUPPLEMENTARY INFORMATION. Supplementary Figures S1-S9. Supplementary Methods

The Human Body: An Overview of Anatomy. Anatomy. Physiology. Anatomy - Study of internal and external body structures

Supplementary Figure 1. Double-staining immunofluorescence analysis of invasive colon and breast cancers. Specimens from invasive ductal breast

Q: In order to use the code 8461/3 (serous surface papillary) for ovary, does it have to say the term "surface" on the path report?

Approved for Public Release; Distribution Unlimited

Supplementary Figures

Supplementary Information POLO-LIKE KINASE 1 FACILITATES LOSS OF PTEN-INDUCED PROSTATE CANCER FORMATION

1. The metastatic cascade. 3. Pathologic features of metastasis. 4. Therapeutic ramifications. Which malignant cells will metastasize?

Choosing Between Lentivirus and Adeno-associated Virus For DNA Delivery

Biochemistry of Cancer and Tumor Markers

Supplementary Figures

Introduction in human anatomy

America, Hershey, PA Australia, Melbourne, VIC Europe, Munich

Supplemental Figure 1

BIT 120. Copy of Cancer/HIV Lecture

Ovarian Tumors. Andrea Hayes-Jordan MD FACS, FAAP Section Chief, Pediatric Surgery/Surgical Onc. UT MD Anderson Cancer Center

Nature Immunology: doi: /ni Supplementary Figure 1. Huwe1 has high expression in HSCs and is necessary for quiescence.

TUMOR,NEOPLASM. Pathology Department, Zhejiang University School of Medicine,

Transcription:

Cancer and Metastasis Reviews 17: 271 277, 1999. 1999 Kluwer Academic Publishers. Printed in the Netherlands. Orthotopic transplant mouse models with green fluorescent protein-expressing cancer cells to visualize metastasis and angiogenesis Robert M. Hoffman AntiCancer, Inc., San Diego, CA, USA; Department of Surgery, UCSD Medical Center, San Diego, CA, USA Key words: metastasis, orthotopic, intact tissue, implantation, GFP, nude mice Summary There have been major efforts in metastasis research in recent years, especially on the role of angiogenesis in the metastatic process. Much of the information in this area has been obtained from model systems that are not representative of clinical cancer. The technique of surgical orthotopic implantation (SOI) has allowed the development of clinically relevant metastatic models of human cancer in immunodeficient rodents such as the nude and SCID mouse. In order to allow direct visualization of the metastatic process, we took advantage of the green fluorescent protein (GFP) of the jellyfish, Aequorea victoria. A series of cancer cell lines have been stably transfected with vectors containing humanized GFP cdna. To utilize GFP expression for metastasis studies, fragments of subcutaneously growing tumor, which were comprised of GFP-expressing cells, were implanted by SOI in nude mice. Subsequent metastases were visualized in systemic organs by GFP fluorescence in the lung, liver, bones, brain and other organs down to the single-cell level. With this fluorescent tool, we detected and visualized for the first time tumor cells at the microscopic level in fresh viable tissue in their normal host organs even in the live animal. Angiogenesis is readily visualized in the transplanted GFP-expressing tumors in real time in situ in the live animal using simple laparotomy and fluorescent techniques. The results with the GFP-transfected tumor cells, combined with the use of SOI, demonstrate a fundamental advance to visualize and study cancer metastasis and the role of angiogenesis and other factors in the metastatic process. Introduction There has been a great interest in the study of metastasis in recent years, especially in the role of angiogenesis in metastasis. Recent results have indicated however, that the choice of animal models is critical for obtaining relevant data on the control of metastasis and the role of angiogenesis and other factors in the metastatic process. Fidler [1] and others have demonstrated the importance of orthotopic transplantation of tumors in appropriate recipients such as nude or SCID mice. The orthotopic site provides the relevant tumors microenvironment [1] for metastasis in contrast to ectopic sites such as the subcutaneous space or even related organs [2]. We have developed the technique of surgical orthotopic implantation (SOI) of histologically intact tumor tissue fragments for clinically relevant metastatic models of human cancer in immunodeficient rodents such as the nude and SCID mouse [3 8]. SOI has been shown by Cowen et al. [9] to be critical for a functional network of vessels to form in liver metastasis from a colon tumor. In contrast, iv injection of tumor cells results in undifferentiated tumor deposits in the lung and liver of the nude mouse without stromal or vascular involvement [9]. The importance of SOI on resulting metastasis and angiogenesis becomes of particular importance with regard to the accurate pre-clinical evaluation of new antiangiogenesis agents whose development thus far has depended on animals that were injected ectopically with suspensions of tumor cells [10 16]. This has been demonstrated in an SOI model of colon cancer where an antiangiogenic agent inhibited colon cancer metastasis but not the primary tumor growth [34]. TYPESET CMR 17.3 98 2 PIPS 195060

272 The early stages of tumor progression and micrometastasis formation have been difficult to analyze due to the inability to identify small numbers of tumor cells against a background of many host cells. For example, the visualization of tumor cell emboli, micrometastases and their progression over real-time during the course of the disease has been difficult to study in current models of metastasis. Previous studies used transfection of tumor cells with the Escherichia coli beta-galactosidase (lacz) gene to detect micrometastases [17,18]. However, detection of lacz requires extensive histological preparation, and therefore it is not possible to detect and visualize tumor cells in viable fresh tissue or the live animal at the microscopic level. The visualization of tumor invasion and micrometastasis formation in viable fresh tissue or the live animal is necessary for a critical understanding of tumor progression and its control. To enhance the resolution of the visualization of micrometastases in fresh tissue, we have utilized the green fluorescent protein (GFP) gene, cloned from the bioluminescent jellyfish Aequorea victoria [19]. GFP has demonstrated its potential for use as a marker for gene expression in a variety of cell types [20,21]. The GFP cdna encodes a 283 amino acid polypeptide with molecular weight of 27 kd [22,23]. The monomeric GFP requires no other Aequorea proteins, substrates, or cofactors to fluoresce [24]. Recently, GFP gene gain-of-function mutants have been generated by various techniques [25 28]. For example, the GFP-S65T clone has the serine-65 codon substituted with a threonine codon which results in a single excitation peak at 490 nm [25]. Moreover, to develop higher expression in human and other mammalian cells, a humanized hgfp-s65t clone was isolated [29]. The much brighter fluorescence in the mutant clones allows for easy detection of GFP expression in transfected cells. We have demonstrated the isolation of stable transfectants of human lung carcinoma cells (ANIP 973 and H-460) and Chinese hamster ovary (CHO) cells that express high-level GFP fluorescence in vitro [30-35] and a series of cell lines from the National Cancer Institute (NCI) cell line screen (H460, colon cancer Colo- 205, prostate cancer PC-3 and DU-145, breast cancer MDA-MB435 and MDA-MB468, kidney cancer SN12C, tongue cancer SCC-25, melanoma LOX). The transfectants are highly fluorescent in vivo in tumors formed from the cells. Using these fluorescent transfectants, orthotopic-transplant animal models [3 8] were utilized for visualizing the metastatic and angiogenic processes in fresh tissue including living animals that heretofore was not possible [30 35]. These developments with GFP-expressing tumor cells provide an invaluable new tool for understanding the most important steps in tumor host-organ interaction, tumor progression, metastasis, and angiogenesis. Materials and methods DNA manipulations and expression vector constructions The dicistronic expression vector (ped-mtx r ) was obtained from Genetics Institute (Cambridge, MA) [33]. The expression vector containing the codonoptimized hgfp-s65t gene was purchased from CLONTECH Laboratories, Inc. (Palo Alto, CA). To construct the hgfp-s65t containing expression vector, phgfp-s65t was digested with Hind III, blunted at the end. The entire hgfp coding region was then excised with Xba I. The ped-mtx r vector was digested with Pst I, blunted at the end, and further digested with Xba I. The hgfp-s65t cdna fragment was then unidirectionally subcloned into ped-mtx r. Cell culture, transfection, selection CHO-K1 cells were cultured in DMEM (GIBCO) containing 10% fetal calf serum (FCS) (Gemini Bioproducts, Calabasas, CA), 2 mm L-gulutamine and 100 µm non-essential amino acids (Irvine Scientific, Santa Ana, CA). ANIP 973 cells and the NCI cell lines were cultured either in MEM (GIBCO), RPMI 1640 (GIBCO) or Ham s F-12 (Irvine Scientific) containing 10% fetal calf serum (FCS) (Gemini Bio-products, Calabasas, CA). For transfection, near-confluent CHO-K1 or ANIP cells were incubated with a precipitated mixture of LipofectAMINE reagent (GIBCO), and saturating amounts of plasmids for 6 h before being replenished with fresh medium. NCI cell lines were transfected with the plein retrovirus containing the EGFP gene (Clontech) using DOTAB liposome transfection reagent (Boerhinger Mannheim). plein expresses EGFP and the neomycin gene on the same bicistronic message [35]. CHO-K1 cells and ANIP were harvested by trypsin/edta 48 h post-transfection, and subcultured at a ratio of 1 : 15 into selective medium which

273 contained various concentrations of methotrexate (MTX) up to 1.5 µm. Cells with stably integrated plasmids were selected by growing transientlytransfected cells in medium with increasing concentrations of MTX. Clones were isolated with cloning cylinders (Bel-Art Products, Pequannock, NJ) with trypsin/edta. They were amplified and transferred with conventional culture methods. CHO Clone-38 and ANIP Clone-26 were chosen because of their highintensity GFP fluorescence and stability. GFP-transfected NCI cell lines were similarly selected but with neomycin (0.5 2 µg/ml) [35]. Surgical orthotopic implantation of CHO-K1 Clone-38 in nude mice Tumor fragments (1 mm 3 ) derived from the nude mouse subcutaneous CHO-K1 Clone-38 tumor were implanted by surgical orthotopic implantation (SOI) on the ovarian serosa in six nude mice [5]. The mice were anesthetized by isofluran inhalation. An incision was made through the left lower abdominal pararectal line and peritoneum. The left ovary was exposed and part of the serosal membrane was scraped with a forceps. Four 1 mm 3 tumor pieces were fixed on the scraped site of the serosal surface with an 8-0 nylon suture (Look, Norwell, MA). The ovary was then returned into the peritoneal cavity, and the abdominal wall and the skin were closed with 6-0 silk sutures. Four weeks later, the mice were sacrificed and the lungs and the other organs were removed. Surgical orthotopic implantation of ANIP 973 human lung cancer in nude mice Tumor pieces (1-mm 3 ) derived from the nude mouse subcutaneous tumor formed from ANIP Clone-26 were implanted by surgical orthotopic implantation (SOI) onto the left visceral pleura in four nude mice [4]. The mice were anesthetized by Isofluran inhalation. A small 1-cm transverse incision was made on the left-lateral chest of the nude mice via the fourth intercostal space. A small incision provided access to the pleural space, and resulted in total lung collapse. Five tumor pieces were sewn together with a 8-0 nylon (Look, Norwell, MA) surgical suture and fixed by making one knot. The lung was taken up by forceps and the tumor sewn into the lower part of the lung with one suture. The lung tissue was then returned into the chest cavity. The chest muscles and skin were closed with a single layer of 6-0 silk sutures. The lung was reinflated by withdrawing air from the chest cavity with a 23-gauge needle. Surgical orthotopic implantation of GFP-transfected PC-3 in the prostate of nude mice was carried out as described [8]. Surgical orthotopic implantation of GFPtransfected H460 was carried out in similar fashion as for ANIP-973 described above. Tumor angiogenesis visualization in live animals with GFP tumors Angiogenesis of GFP tumors was visualized in live animals by injection of 200 µl of0.1µm rhodamine five minutes before surgical exploration. A GFP filter set with an excitation band-pass filter (D425/60) and for emission, a long pass filter (GG475) (Chroma Technology, Brattleboro, UT) was used for fluorescence with a Leica Stereo Microscope. Results and discussion Isolation of stable high-level expression GFP transfectants of CHO-K1 and ANIP cells The expression-vector transfected CHO-K1 and ANIP cells were able to grow in levels of MTX up to 1.5 µm. The selected MTX resistant CHO and ANIP cells had a striking increase in GFP fluorescence compared to the transiently-transfected cells. A subclone was termed CHO-K1 GFP 38 (Clone-38) which was stable in 1.5 µm MTX, possibly due to chromosomal integration of the amplified GFP genes [30]. There was no difference in the cell proliferation rates of parental cells and selected transfectants determined by comparing their doubling times. A subclone of ANIP 973 which expressed the strongest GFP was isolated and termed ANIP 973-hGFP-S65T-Clone-26 (Clone 26) [31]. Patterns of metastasis of ovarian tumor after SOI visualized by GFP Nude mice were implanted with 1-mm 3 cubes of CHO- K1 Clone-38 tumor into the ovary and were sacrificed at four weeks [30]. All mice had tumors in the ovaries. The tumor had also seeded throughout the peritoneal cavity, including the colon, cecum, small intestine, spleen, and peritoneal wall. The primary tumor and peritoneal metastases were strongly fluorescent. Numerous micrometastases were detected by fluorescence on the

274 Figure 1. Ribmetastasis of GFP-expressing human prostate PC-3 tumor (arrow) after surgical orthotopic transplantation (Bar = 200 µm). Figure 2. Metastasis of GFP-expressing human lung cancer human lung H460 tumor to the spine after SOI to a nude mouse (Bar = 325 µm). Figure 3. Angiogenesis visualized in GFPexpressing ANIP-973 human lung tumor cells growing on the wall of the nude mouse colon after injection of rhodamine dye (see Materials and methods for detail). Thick arrows denote perimeter of tumor. Thin arrows denote tumor blood vessels. (Bar = 200 µm).

275 lungs of all mice. Multiple micrometastasis were also detected by fluorescence on the liver, kidney, contralateral ovary, adrenal gland, para-aortic lymph node, and pleural membrane down to the single-cell level. Singlecell micrometastases could not be detected by standard histological techniques. Even these multiple-cell small colonies were difficult to detect by hematoxylin and eosin staining, but could be detected and visualized clearly by GFP-fluorescence. Patterns of metastasis of lung tumor after SOI visualized by GFP expression Primary tumors grew in the operated left lung in all mice after SOI of GFP-transfected ANIP 973 Clone-26. GFP expression allowed visualization of the advancing margin of the tumor spreading in the ipsilateral lung. All animals explored had evidence of chest wall invasion and local and regional spread. Metastatic contralateral tumors involved the mediastinum, contralateral pleural cavity, the contralateral visceral pleura, occurring in all mice. While, the ipsilateral tumor had a continuous and advancing margin, the contralateral tumor seemed to have been formed by multiple seeding events. These observations were made possible by GFP fluorescence of the fresh tumors tissue [31,32]. When non-gfp-transfected ANIP was compared with GFP-transformed ANIP for metastatic capability similar results were seen [31]. Contralateral hilar lymph nodes were also involved shown by GFP expression. A cervical lymph node metastasis was brightly visualized by GFP in fresh tissue [31,32]. GFP-PC-3 transplanted by SOI in the prostate demonstrated extremely high malignancy with seeding in the liver, lung, skull, bones (Figure 1) and brain [35]. GFP H460 transplanted by SOI in the lung demonstrated extensive bone metastasis throughout the skeleton (Figure 2) and metastasis in many other organs [35]. To study tumor progression and the metastatic process at the earliest stages, it was important to use a metastatic model which could allow spontaneous metastasis in the animal. In the last 10 years, it has become clear that orthotopic sites of implantation are critical for the metastatic expression of transplanted tumors in nude mice. We have found that the use of tissue fragments rather than dispersed cells allows full expression of the metastatic capability of surgically orthotopically implanted (SOI) tumors [3 8]. GFPgene-transfected CHO-K1 Clone-38 cells were successfully used to visualize extensive peritoneal seeding as well as distant macro- and micro-metastases in nude mice including the lung following SOI in the ovary of subcutaneous tumor fragments. We utilized the SOI model and GFP-expressing ANIP Clone-26 tumor to visualize human lung tumor growth and metastases to the mediastinum and contralateral lung, as well as to lymph nodes. PC-3 and H460 have high bone metastatic capability as visualized with GFP. These are very important clinical events and can now be visualized much more realistically by GFP in fresh tissue. These models provided the opportunity to demonstrate the capability of GFP for detection and visualization of metastases. We have visualized actively colonizing as well as dormant tumor cells in the lung and brain. Dormant micrometastasis is one of the most important steps to understand in tumor progression [34]. In recent studies, the mechanism of this important phenomenon was studied with regard to angiogenesis and other chemical regulators of tumor colonization [34,10 16]. However, these experimental models did not allow direct observation of the dormant colonies in fresh live tissue or the live animal as can be done with GFP-expressing tumor cells. The GFP-expressing tumors have been shown to be useful for visualization of early to late stage angiogenesis in the living animal (Yang and Hoffman, unpublished results). Each tumor deposit can be observed for extent of angiogenesis over time by surgical exploration with fluorescence microscopy (Figure 3). Results thus far suggest site and tumor type influence the extent and apparent dependence on angiogenesis for tumor expansion (Yang and Hoffman, unpublished results). The methods described in this review, SOI and GFP expression, will facilitate the understanding of tumor growth and progression including seeding and target organ colonization and the role of angiogenesis in this process. These studies should provide new insights into metastatic mechanisms, and evaluation of new agents for treatment of metastatic disease. References 1. Fidler IJ: Critical factors in the biology of human cancer metastasis. Cancer Res 50: 6130 6138, 1990 2. Togo S, Shimada H, Kubota T, Moossa AR, Hoffman RM: Host organ specifically determines cancer progression. Cancer Res 55: 681 684, 1995

276 3. Astoul P, Colt HG, Wang X, Hoffman RM: A patient-like nude mouse model of parietal pleural human lung adenocarcinoma. Anticancer Res 14: 85 92, 1994 4. Wang X, Fu X, Hoffman RM: A new patient-like metastatic model of human lung cancer constructed orthotopically with intact tissue via thoracotomy in immunodeficient mice. Int J Cancer 51: 992 995, 1992 5. Fu X, Hoffman RM: Human ovarian carcinoma metastatic models constructed in nude mice by orthotopic transplantation of histologically-intact patient specimens. Anticancer Res 13: 283 286, 1993 6. Hoffman RM: Orthotopic is orthodox: Why are orthotopictransplant metastatic models different from all other models? J Cellular Biochem 56: 1 3, 1994 7. Kuo T, Kubota T, Watanabe M, Furukawa T, Teramoto T, Ishibiki K, Kitajima M, Moossa AR, Penman S, Hoffman RM: Liver colonization competence governs colon cancer metastasis. Proc Natl Acad Sci USA 92: 12085 12089, 1995 8. An Z, Wang X, Geller J, Moossa AR, Hoffman RM: Surgical orthotopic implantation allows high lung and lymph node metastatic expression of human prostate carcinoma cell line PC-3 in nude mice. The Prostate 34: 169 174, 1998 9. Cowen SE, Bibby MC, Double JA: Characterisation of the vasculature within a murine adenocarcinoma growing in different sites to evaluate the potential of vascular therapies. Acta Oncologica 34: 357 360, 1995 10. O Reilly MS, Holmgren L, Shing Y, Chen C, Rosenthal RA, Moses M, Lane WS, Cao Y, Sage EH, Folkman J: Angiostatin: A novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma. Cell 79: 315 328, 1994 11. O Reilly MS, Holmgren L, Chen C, Folkman J: Angiostatin induces and sustains dormancy of human primary tumors in mice. Nature Med 2: 689 692, 1996 12. Folkman J: Angiogenesis in cancer, vascular, rheumatoid and other diseases. Nature Med 1: 27 31, 1995 13. O Reilly MS, Boehm T, Shing Y, Fukai N, Vasios G, Lane WS, Flynn E, Birkhead JR, Olsen BR, Folkman J: Endostatin: An endogenous inhibitor of angiogenesis and tumor growth. Cell 88: 277 285, 1997 14. Boehm T, Folkman J, Browder T, O Reilly MS: Antiangiogenesis therapy of experimental cancer does not induce acquired drug resistance. Nature 390: 404 407, 1997 15. Arap W, Pasqualini R, Ruoslahti E: Cancer treatment by targeted drug delivery to tumor vasculature in a mouse model. Science 279: 377 380, 1998 16. Pasqualini R, Koivunen E, Ruoslahti E: αv integrins as receptors for tumor targeting by circulating ligands. Nature Biotech 15: 542 546, 1997 17. Lin WC, Pretlow TP, Pretlow TG, Culp LA: Bacterial lacz gene as a highly sensitive marker to detect micrometastasis formation during tumor progression. Cancer Res 50: 2808 2817, 1990 18. Lin WC, Culp LA: Altered establishment/clearance mechanisms during experimental micrometastasis with live and/or disabled bacterial lacz-tagged tumor cells. Invasion Metas 12: 197 209, 1992 19. Morin J, Hastings J: Energy transfer in a bioluminescent system. J Cell Physiol 77: 313 318, 1972 20. Chalfie M, Tu Y, Euskirchen G, Ward WW, Prasher DC: Green fluorescent protein as a marker for gene expression. Science 263: 802 805, 1994 21. Cheng L, Fu J, Tsukamoto A, Hawley RG: Use of green fluorescent protein variants to monitor gene transfer and expression in mammalian cells. Nature Biotech 14: 606 609, 1996 22. Prasher DC, Eckenrode VK, Ward WW, Prendergast FG, Cormier MJ: Primary structure of the Aequorea victoria green-fluorescent protein. Gene 111: 229 233, 1992 23. Yang F, Miss LG, Phillips GN Jr: The molecular structure of green fluorescent protein. Nature Biotech 14: 1252 1256, 1996 24. Cody CW, Prasher DC, Welstler VM, Prendergast FG, Ward WW: Chemical structure of the hexapeptide chromophore of the Aequorea green fluorescent protein. Biochemistry 32: 1212 1218, 1993 25. Heim R, Cubitt AB, Tsien RY: Improved green fluorescence. Nature 373: 663 664, 1995 26. Delagrave S, Hawtin RE, Silva CM, Yang MM, Youvan DC: Red-shifted excitation mutants of the green fluorescent protein. Bio/Technology 13: 151 154, 1995 27. Cormack B, Valdivia R, Falkow S: FACS-optimized mutants of the green fluorescent protein (GFP). Gene 173: 33 38, 1996 28. Cramer A, Whitehorn EA, Tate E, Stemmer WPC: Improved green fluorescent protein by molecular evolution using DNA shuffling. Nature Biotech 14: 315 319, 1996 29. Zolotukhin S, Potter M, Hauswirth WW, Guy J, Muzycka N: Humanized green fluorescent protein cdna adapted for high-level expression in mammalian cells. J Virology 70: 4646 4654, 1996 30. Chishima T, Miyagi Y, Wang X, Yamaoka H, Shimada H, Moossa AR, Hoffman RM: Cancer invasion and micrometastasis visualized in live tissue by green fluorescent protein expression. Cancer Research 57: 2042 2047, 1997 31. Chishima T, Miyagi Y, Wang X, Baranov E, Tan Y, Shimada H, Moossa AR, Hoffman RM: Metastatic patterns of lung cancer visualized live and in process by green fluorescence protein expression. Clin Exper Metas 15: 547 552, 1997 32. Chishima T, Miyagi Y, Wang X, Tan Y, Shimada H, Moossa AR, Hoffman RM: Visualization of the metastatic process by green fluorescent protein expression. Anticancer Res 17: 2377 2384, 1997 33. Kaufman RJ, Davies MV, Wasley LC, Michnick D: Improved vectors for stable expression of foreign genes in mammalian cells by use of the untranslated leader sequence from EMC virus. Nucleic Acids Res 19: 4485 4490, 1991 34. Holmgren L, O Reilly MS, Folkman J: Dormancy of micrometastases: Balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nature Med 1: 149 153, 1995 35. Yang M, Hasegawa S, Jiang P, Wang X, Tan Y, Chishima T, Shimada H, Moossa AR and Hoffman RM: Widespread skeletal metastatic potential of human lung cancer revealed by green fluorescent protein expression. Cancer Res 58: 4217 4221, 1998

277 36. Tanaka T, Konnott, Matsuda I, Nakamura S, and Baba S: Prevention of hepatic metastasis of human colon cancer by angiogenesis inhibition TNP-470. Cancer Res 55: 836 839, 1995 Address for offprints: Robert M. Hoffman, AntiCancer, Inc., 7917 Ostrow Street, San Diego, CA 92111. Tel: 619-654-2555; Fax: 619-268-4175; e-mail: all@anticancer.com