+ All Categories
Home > Documents > Tumor Heterogeneity and the Biology of Cancer Invasion and ......Tumor Heterogeneity and the Biology...

Tumor Heterogeneity and the Biology of Cancer Invasion and ......Tumor Heterogeneity and the Biology...

Date post: 25-Jan-2021
Category:
Upload: others
View: 3 times
Download: 1 times
Share this document with a friend
11
[CANCERRESEARCH38,2651-2660,September1978] Abstract The development of a metastasis Is dependent on an interplay between host factors and intrinsic character istics of malignant tumor cells. The process of metastasis is highly selective, and the metastatic lesion represents the end point of many destructive events that only a few cells can survive. Neoplasms, which are predominantly heterogeneous, contain a variety of subpopulatlons of cells with differing metastatic potential. Furthermore, metastatic cell variants have been shown to preexist in murine neoplasms of old and recent origin. The possible existence of highly metastatic variant cells within a pri mary tumor suggests that we no longer should consider a neoplasm to be a uniform entity. Efforts to design effective therapeutic agents and procedures against malignant tumors should be directed toward the few but fatal metastatic subpopulations of cells. Introduction Metastasis is defined by Dorland (8) as the transfer of disease from one organ or part to another not directly connected with it. The metastasis of cancer cells is one of the most devastating aspects of neoplasia; it is responsible for most therapeutic failures because patients succumb to multiple secondary tumor growths and not necessarily to the primary tumor. Major advances in surgical techniques, coupled with advances in general patient care, have in creased the success of the resection of primary neoplasms. However, in the majority of patients with clinical cancer, excluding those with skin tumors, metastasis has already occurred at the time of diagnosis (53, 58, 59). Therefore, even the most extensive surgical procedures cannot hope to bring about a high â€oecure― rate. Short of the complete prevention of cancer, the urgent goal of the oncologist should be the prevention or the successful treatment of micrometastases. Additional approaches to the therapy of disseminated disease may be forthcoming if the underlying pathogenesis of cancer metastasis can be clearly understood. The out come of the metastatic process depends on the properties of both host and tumors, and the balance of these contri butions probably varies in tumor systems (15, 58). Animal models have proven invaluable in the elucidation of the host and tumor factors involved in determining the outcome of cancer metastasis. The proper utilization of such models may advance considerably our understanding I Research sponsored by the National Cancer Institute under Contract NO1-CO-75380 with Litton Bionetics, Inc. Presented as the lecture on Advancesin Oncologyat the 69thAnnualMeetingof the AmericanAssocia tionfor CancerResearch,April 1978,Washington,D.C. Received May 24, 1978; accepted June 5, 1978. of the biology of the phenomenon and, therefore, allow the development of new approaches to the therapy of dissemi nated disease. We have recently developed one such animal model for qualitative studies of metastasis. Mice are given i.d. injec tions, in the external ear, of 0.05 ml of tumor cell suspen sion. Three to 4 weeks later, when the tumors are estab lished, the ear is amputated at its base and the mice are allowed to survive. Six to 8 weeks later, the mice are killed and examined for the presence of lymph node or visceral metastases. In the model shown in Fig. la, 25,000 viable cells of the B16 melanoma, syngeneic to the C57BL/6 mouse, were injected into the medial surface of the external ear. The ear and growing tumor were amputated 3 weeks after s.c. injection. If the tumor mass contained no meta static cells, the amputation of the ear would be curative. Alternatively, if the growing tumor contained some meta static cells, which invaded blood vessels and lymphatics prior to amputation of the ear, the mice would eventually die of distant tumor growths. As seen in Fig. lb, in some mice growth of tumor occurred in the lymph nodes regional to the ear, i.e., the cervical and submandibular nodes, and in the lungs. The components of this qualitative assay for the metastatic potential of murine neoplasms are shown in Fig. lc. This model or procedure may also be useful for determining the metastatic potential of murine neoplasms in studies that evaluate the therapy of spontaneous metas tases. Pathogenesis of Metastasis The first steps in the pathogenesis of metastasis (Chart 1) are the local invasion of normal host tissues, penetration into lymphatics or blood vessels by malignant cells, and detachment. The actual mechanism for tumor cell invasion remains unclear; it probably depends on both tumor and hostfactors. Tumor cells can spread by three major routes. The first involvespreadbydirectextensioninwhichatumorgrow ing in a body cavity releases cells or fragments that can seed serosal and/or mucosal surfaces and develop into new growths. Two examples are lung mediastinal tumors that enter the pleural cavity and malignant ovarian tumors that shed cells into the peritoneal cavity. Primary tumors of the central nervous system, although highly invasive, rarely produce metastases in organs outside the nervous system. The mode of their spread appears to be by direct extension or via the cerebrospinal fluid. The second and third routes of spread, which are the subject of this lecture, are via the lymphatic and hematogenous compartments of the circula tory system. Clinical observations have suggested that carcinomas frequently spread and grow in the lymphatic system, whereas malignant tumors of mesenchymal origin spread SEPTEMBER 1978 2651 Tumor Heterogeneity and the Biology of Cancer Invasion and Metastasis1 Isaiah J. Fidler Cancer Biology Program, National Cancer Institute Frederick Cancer Research Center, Frederick, Maryland 21701 on May 31, 2021. © 1978 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from
Transcript
  • [CANCERRESEARCH38,2651-2660,September1978]

    Abstract

    The development of a metastasis Is dependent on aninterplay between host factors and intrinsic characteristics of malignant tumor cells. The process of metastasisis highly selective, and the metastatic lesion representsthe end point of many destructive events that only a fewcells can survive. Neoplasms, which are predominantlyheterogeneous, contain a variety of subpopulatlons ofcells with differing metastatic potential. Furthermore,metastatic cell variants have been shown to preexist inmurine neoplasms of old and recent origin. The possibleexistence of highly metastatic variant cells within a primary tumor suggests that we no longer should consider aneoplasm to be a uniform entity. Efforts to designeffective therapeutic agents and procedures againstmalignant tumors should be directed toward the few butfatal metastatic subpopulations of cells.

    Introduction

    Metastasis is defined by Dorland (8) as the transfer ofdisease from one organ or part to another not directlyconnected with it. The metastasis of cancer cells is one ofthe most devastating aspects of neoplasia; it is responsiblefor most therapeutic failures because patients succumb tomultiple secondary tumor growths and not necessarily tothe primary tumor. Major advances in surgical techniques,coupled with advances in general patient care, have increased the success of the resection of primary neoplasms.However, in the majority of patients with clinical cancer,excluding those with skin tumors, metastasis has alreadyoccurred at the time of diagnosis (53, 58, 59). Therefore,even the most extensive surgical procedures cannot hopeto bring about a high “cure―rate. Short of the completeprevention of cancer, the urgent goal of the oncologistshould be the prevention or the successful treatment ofmicrometastases.

    Additional approaches to the therapy of disseminateddisease may be forthcoming if the underlying pathogenesisof cancer metastasis can be clearly understood. The outcome of the metastatic process depends on the propertiesof both host and tumors, and the balance of these contributions probably varies in tumor systems (15, 58).

    Animal models have proven invaluable in the elucidationof the host and tumor factors involved in determining theoutcome of cancer metastasis. The proper utilization ofsuch models may advance considerably our understanding

    I Research sponsored by the National Cancer Institute under Contract

    NO1-CO-75380 with Litton Bionetics, Inc. Presented as the lecture onAdvancesin Oncologyat the 69thAnnualMeetingof the AmericanAssociation for CancerResearch,April 1978,Washington,D. C.

    Received May 24, 1978; accepted June 5, 1978.

    of the biology of the phenomenon and, therefore, allow thedevelopment of new approaches to the therapy of disseminated disease.

    We have recently developed one such animal model forqualitative studies of metastasis. Mice are given i.d. injections, in the external ear, of 0.05 ml of tumor cell suspension. Three to 4 weeks later, when the tumors are established, the ear is amputated at its base and the mice areallowed to survive. Six to 8 weeks later, the mice are killedand examined for the presence of lymph node or visceralmetastases. In the model shown in Fig. la, 25,000 viablecells of the B16 melanoma, syngeneic to the C57BL/6mouse, were injected into the medial surface of the externalear. The ear and growing tumor were amputated 3 weeksafter s.c. injection. If the tumor mass contained no metastatic cells, the amputation of the ear would be curative.Alternatively, if the growing tumor contained some metastatic cells, which invaded blood vessels and lymphaticsprior to amputation of the ear, the mice would eventuallydie of distant tumor growths. As seen in Fig. lb, in somemice growth of tumor occurred in the lymph nodes regionalto the ear, i.e., the cervical and submandibular nodes, andin the lungs. The components of this qualitative assay forthe metastatic potential of murine neoplasms are shown inFig. lc. This model or procedure may also be useful fordetermining the metastatic potential of murine neoplasmsin studies that evaluate the therapy of spontaneous metastases.

    Pathogenesis of Metastasis

    The first steps in the pathogenesis of metastasis (Chart 1)are the local invasion of normal host tissues, penetrationinto lymphatics or blood vessels by malignant cells, anddetachment. The actual mechanism for tumor cell invasionremains unclear; it probably depends on both tumor andhostfactors.

    Tumor cells can spread by three major routes. The firstinvolvesspreadbydirectextensioninwhich a tumor growing in a body cavity releases cells or fragments that canseed serosal and/or mucosal surfaces and develop into newgrowths. Two examples are lung mediastinal tumors thatenter the pleural cavity and malignant ovarian tumors thatshed cells into the peritoneal cavity. Primary tumors of thecentral nervous system, although highly invasive, rarelyproduce metastases in organs outside the nervous system.The mode of their spread appears to be by direct extensionor via the cerebrospinal fluid. The second and third routesof spread, which are the subject of this lecture, are via thelymphatic and hematogenous compartments of the circulatory system.

    Clinical observations have suggested that carcinomasfrequently spread and grow in the lymphatic system,whereas malignant tumors of mesenchymal origin spread

    SEPTEMBER 1978 2651

    Tumor Heterogeneity and the Biology of Cancer Invasion and Metastasis1

    Isaiah J. Fidler

    Cancer Biology Program, National Cancer Institute Frederick Cancer Research Center, Frederick, Maryland 21701

    on May 31, 2021. © 1978 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • I. J. Fidler

    Primary MalignantNeoplasm

    Vascularization

    Interactionwith HostPlatelets,LymphocytesandOther BloodElements

    J

    LymphaticsVenulesCapIllaries

    Chart 1. The pathogenesis of a metastasis. The outcome of the sequential steps ofthe process depends on the interaction oftumor cellswith their host.

    Arrestin CapIllaryBedofOrgans

    Lung

    Metastases

    more frequently via the hematogenous route (7). These twocategories are rather arbitrary, however, because the bloodand lymph systems are intimately interlinked. The studiesof del Regato (7), Fisher and Fisher (18-20), Wood (60),Zeidman (61, 62), and Zeidman and Buss (63) clearly demonstrated that cancer cells may invade the lymphatics directly or may gain access to them via blood vessels and thatcancer cells that invade lymphatics can find their way toblood vessels via venolymphatic anastomoses or by way ofthe thoracic duct.

    The thin-walled venules, like the lymphatic channels,offer little resistance to penetration by tumor cells and thusprovide the most common pathway for the entry of tumorcells into the circulation. In contrast, the arteries, the wallsof which contain elastic and collagen fibers, are rarelyinvaded by tumor cells. After infiltrating the vessels, tumorcells can detach and be carried away passively in thebloodstream or remain localized and proliferate at the siteof vessel invasion. Frequently, a thrombus will form aroundactively growing tumor cells that have penetrated the circulatory system , and it has been suggested by many investigators (1, 3—5)that a direct correlation exists betweenthegrowth potential of a malignant tumor and the formation offibrin around it.

    Detachment and embolization of tumor cells, regardlessof whether their transport is via the lymphatics or blood,are probably continuous processes (56, 57). Most malignanttumors have a well-established blood supply with multiplethin-walled vessels, and Franks (22) has suggested that thedevelopment of metastases begins immediately with thegrowth of primary malignant cancers. In support he citesthe large number of clinical cases in which primary tumorswere surgically removed, and yet the patient succumbed tometastatic lesions many years later. A sudden change in

    venous pressure, such as that occurring during a cough,could lead to momentary blood turbulence and the releaseof a shower of emboli (61). Similarly, diagnostic proceduresand surgical trauma may cause a sudden increase in thenumber of tumor cells released into the circulation (20).

    However, the presence of tumor cells in the circulationdoes not always lead to a metastasis (15, 47, 53, 58). In anextensive reviewof the literature on circulating tumor cells,Salsbury (47) concluded that there is no evidence that themere presenceof tumor cells in the circulation indicates aworse prognosis than the absenceof tumor cells.

    During circulatory transport, tumor cells can undergo avariety of interactions, including aggregation with othertumor cells (39), platelets (24), lymphocytes, and other hostcells (14, 39). Some tumor cells are thromboplastic andelicit fibrin formation either during their circulation or soonafter their arrest in capillary beds (1, 3, 55, 60). If bloodborne tumor cells are aggregated by homotypic or heterotypic cell interactions or by soluble blood components intolarge emboli, their success in forming tumors after theirarrest in the microcirculation should be increased (35).

    It has been demonstrated experimentally that larger emboli are more effective per input tumor cell in implantationand in survival to form gross tumor colonies after i.v.injection (12).2 Thus, purely mechanical factors such asembolic size and deformability (as well as capillary diameterand deformability) should be important in the implantationprocess (48). The rates at which tumor cells or their cellemboli pass through capillary beds are not related to cell oremboli size but instead appear to be related to their deform

    2 M. L. Kripke, E. Gruys, and I. J. Fidler. Metastatic Heterogeneity of Cells

    from an UltravioletLight-inducedMurine Fibrosarcomaof RecentOrigin,submitted for publication.

    CANCERRESEARCHVOL. 382652

    Invasion Transport

    Extravasatlon AdherenceofTumor Cells

    EstablIshmentofMlcreenvlronmentandGrowthInto

    on May 31, 2021. © 1978 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • Tumor Heterogeneity and Metastases

    were adapted to tissue culture. The B16 variants, whichgrew in culture after the first in vivo selection, were established as a continuous line and were designated B16-F1.Cells from this line were reinjected into new syngeneicanimals, and 3 weeks later a new group of lung tumorcolonies was removedand cultured to yield line B16-F2andso on. With each succeeding cycle in vivo, the ability of theselected B16 linesto implant, survive, and form lung tumorsincreased (13). After 10 such selections B16-F10 was obtamed, which forms significantly more gross lung tumorsper input cell than does B16-F1 after i.v. or i.c.3 injectioninto syngeneic mice (17).

    Clinical observations of a large number of patients withtumors of defined histological classification suggest thatthere is a tendency for primary tumors arising in an organto metastasize to and grow in particular distant organs (59).In many experimental tumor systems, selective patterns ofhoming and subsequent growth into metastases have beendemonstrated. For example, as early as 1947 Cloudman (6)showed differences betweenthe homing patterns of murinelymphomas and plasmacytomas. Murine thymomas tend tometastasize to and grow in all the lymphoid organs, buttransplantable plasmacytomas tend to metastasize to thebone marrow (43-45). Other tumors that have been reportedto demonstrate organ specificity for metastasis are melanomas (17, 26, 30), histiocytomas (9), and fibrosarcomas(52).

    Thus, clinical and experimental observations of the predilection of certain neoplasms for growth in specific organssuggest that the pattern of metastasis is not random tumorcell distribution but rather that it reflects properties of thecirculating malignant cells, host capillary endothelium, andorgan environment. Two long-standing arguments havebeen advanced to explain the distinctive patterns of distribution of metastases. In 1928 Ewing (10) initially suggestedthat metastasis is influenced by purely “mechanicalfactors,―such as anatomic and hemodynamic factors in thevasculature. In contrast, in 1889 Paget (42) suggested the“seedand soil―hypothesis, which states that the microenvironment of one organ may favor the arrest and subsequent growth of tumor emboli, while the microenvironmentof another organ may not.

    More recently, a third hypothesis has emerged that proposes that properties of the tumor cells themselves mayalso influence their patterns of spread and growth. Aremarkable demonstration of tumor cells homing to andgrowing in specific organs was first reported by Kinsey(30),who used the Cloudman melanoma, and then by Sugarbaker et a!. (52), who used a murine fibrosarcoma. In theirexperiments the tumors shown to spread from a s.c. site tothe lungs were implanted s.c. The tumor cells metastasizednot only to the lung but also to small fragments of lungtissue implanted s.c. They did not metastasize to otherorgan fragments implanted s.c. , which served as controls.

    In the B16 system, the ability of B16-F10 but not theoriginal B16-FO to form lung metastases exclusively aftereither i.v. or i.c injection, even in parabiotically joinedanimals, suggested that the selection of tumor cells withpreferencefor a particular organ might be possible. Recent

    aTheabbreviationusedis:ic., intracardial.

    ability during transcapillary transport (62, 63). Under certainconditions, the adhesion of tumor cells to endothelium insusceptible organs could lead to vessel wall damage andthe subsequent accumulation of neutrophils. Since leukocytes commonly passthrough the endothelium, tumor cellscould enter the extravascular tissues by following pathwaysset by leukocytes that have traversed the vessel wall (55).Alternatively, platelets can aggregate at the site of tumorcell lodgement and release mediators that could contributeto vascular spasm, increased endothelial permeability and,perhaps, increased motility of tumor cells (28).

    The process of extravasation may be facilitated by aspecific tumor cell product(s) that is chemotactic to othertumor cells but not to normal cells (41). When malignantcells reach an extravascular environment, they usually continue to proliferate (Fig. 2). In malignant lesions, vascularization of the micrometastases is probably enhanced by atumor angiogenesis factor, a glycoprotein that stimulatesendothelial cell movement and division (21).

    Metastasis as a Selective Process

    It is not possible to give an all-inclusive review of the vastliterature on the clinical and experimental aspects of cancermetastasis. Therefore, I have chosen to address only onemajor issue which has recently received a great deal ofattention and which I feel has far-reaching consequencesfor our understanding of cancer biology in general and forour approaches to the therapy of disseminated disease inparticular.

    Specifically, I wish to discuss the possibility that neoplasms are heterogeneous and contain subpopulations ofcells with differing metastatic capabilities. We can ask,“Doesthe process of metastasis represent the randomsurvival of tumor cells, or does it result from the survivaland growth of a specialized subpopulation of cells?―Beforeproceeding to consider this question, I shall give you thehistorical background that led to its formulation.

    Several years ago we investigated the fate of circulatingtumor emboli following the i.v. injection of [1251J-5-iodo-2'-deoxyuridine-labeled tumor cells. This technique allows usto determine the distribution and survival of tumor cells intheir recipient. Injected mice were killed at different times,and their organs were collected and processedto determinethe number of viable radioactive tumor cells. The majorityof the injected tumor cells were arrested initially in thelungs. Tumor cell death began shortly thereafter. By 24 hrafter injection, only 1% of the injected tumor cells hadsurvived in host organs, and by Day 14, at which time tumorcolonies were visible in the lungs, less than 0.1% of theoriginal cells injected had survived (11). The fact that themajority of the circulating emboli died and only a minoritysurvived to yield metastases was not surprising but raisedthe following question. Did the 0.1% of the emboli surviveat random or did the surviving cells represent a subpopulation of cells with properties that enhanced their survival andgrowth?

    To answer this question we performed the followingexperiment. The B16 melanoma was injected i.v. into syngeneic C57BL/6 mice. Three weeks later pulmonary tumorcolonies were dissected free of lung tissue, and tumor cells

    SEPTEMBER1978 2653

    on May 31, 2021. © 1978 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • Table 1Heterogeneity of the B16 melanoma parent tumor for experimental metastasis

    Median no. of pulmo- No. of animals with extrapulmoSource of cells° nary metastases nary metastases

    I. J. Fidler

    experiments by Brunson et a!. (2) have clearly demonstratedsuch selection. In their studies a brain-preferring B16 melanoma linewas selectedby repeatedcyclingin vivoofmelanoma cells obtained from the rare brain metastasesthat developed initially following the i.c. injection of theparent tumor. Similarly, Nicolson and Brunson (38) haverecently reported the selection of tumor variants that canhome to and grow in the adrenal gland or ovary afterapproximately l2in vivo selections. These selected variantsare of immense importance, for they may allow us toidentify the determinants responsible for the preference oftumor growth for a particular organ. Collectively, the abovedata suggest that the survival of a few tumor cells thatsubsequently developed into metastases was not a randomoccurrence but was due to certain unique properties of thesurviving cells.

    Heterogeneity of Murine Malignant Neoplasms

    What remained unanswered was the question of whetherthese unique metastatic cells preexisted in the tumor cellpopulation or whether they arose during metastasis by aprocess of adaptation to local environmental conditions. Ifhighly metastatic variant cells could be shown to preexist inthe parent population, this would support the suggestionby Nowell (40) that tumor cell variants arise within developing tumors, are subjected to host selection pressures, andare responsible for the emergence of new sublines withincreased malignant potential.

    To distinguish between these possibilities, my colleagueMargaret Kripke and I performed an experiment similar indesign to the classical fluctuation test devised by Luria andDelbrück (36) to distinguish between selection and adaptation in the origin of bacterial mutants. In our study a cellsuspension of the B16 melanoma parent line was divided

    into two parts. One portion was used for i.v. injection intosyngeneic C57BL/6 mice. The other portion was used toproduce 17 clones, which were then also injected i.v. intogroups of C57BL/6 mice. Eighteen days after the tumorcells were injected, the number of lung metastases in eachrecipient was counted. If the number of metastatic foci inthe lungs of the mice receiving the cloned sublines wassimilar to the number of foci seen in mice receiving theparent line, this would indicate that the parent populationwas homogeneous and that the metastatic foci probablyresulted from adaptation during the process of metastasis.Alternatively, if the cloned sublines gave rise to widelydifferent numbers of lung colonies, this would suggest thatthe parent tumor was heterogeneous and that cells of bothhigh and low metastatic potential preexisted in the parentpopulation.

    As shown in Table 1, the study clearly demonstrated thatthe B16 melanoma is heterogeneous and that a few highlymetastatic tumor cell variants preexisted in the parentalpopulation. There was also considerable variation amongthe clones in the number and sites of pulmonary metastasesproduced following i.v. injection. Control subcloning experiments demonstrated that the variability among theclones was not generated during the cloning procedure(16). The extreme degree of heterogeneity observed withthe B16 melanoma is not surprising considering that thetumor arose in 1954 and has been transplanted for approximately 10 times the life span of a mouse. Repeated passages, both in animals and in cell culture, have providedample opportunity for variant cell types to arise. For thisreason we wished to determine whether a tumor of a morerecent origin would also exhibit heterogeneity by the critenon of metastatic potential. To address this question weused a fibrosarcoma induced in a C3H mouse by chronicUV irradiation (32). Following the s.c. injection of cells, this

    40.5 (8-131)@'B16 parent line(60)b

    Clone 16(10)Clone15(11)Clone 12(9)Clone24 (9)Clone 19(10)Clone7 (10)Clone21 (8)Clone 18(11)Clone 5 (10)Clone6 (9)Clone 17(9)Clone3 (9)Clone 1 (9)Clone 2(10)Clone 13(9)Clone 14 (9)Clone9 (10)

    8/60 ovary, 11/60 lymph nodes, 6/60 liver, 4/60 kidney,3/60 gut, 2/60 adrenal

    0/101/11 lymph node0/91/9 ovary, 1/9 liver, 1/9 lymph node0/100/101/8 lymph node0/110/100/90/91/9 lymph node0/90/102/9 ovary, 1/9 liver2/9 ovary6/10 lymph node, 2/10 adrenal, 1/

    10 kidney, 2/10 liver

    3.556

    101317183645.599

    150214237254.5260

    >500>500

    (2-15)(2-20)(0-34)(5-29)(0-42)(0-43)(1-48)(0-91)(2-171)(5—232)

    (104—210)(160-450)

    (73-321)(7-450)

    (50-350)

    a C57BL/6miceweregiveninjectionsin the tail veinof 50,000viablesinglecellsandkilled 18 days later. The number of pulmonary tumor colonies was determined with theaid of a dissecting microscope.

    b Numbers in parentheses, number of mice per group.C Numbers in parentheses, range.

    2654 CANCERRESEARCHVOL. 38

    on May 31, 2021. © 1978 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • Heterogeneityofthe UV-2237fibrosarcomaparenttumorforexperimentalmetastasisSourceof

    ceIls@'Medianno. of pulmo

    nary metastasesNo.of extrapulmonarymetastasespbUV-2237

    parentline (34)(@1

    60.5(17-300)@'Ø@i34Clone

    15(10)1(0-9)0/10

  • Table 3Spontaneousmetastasisof UV-2237parent tumor and its clone

    subpopulations

    Growth of s.c. tumors@@No.of mice

    with metasta- Av. sizeat 2 wkSourceof cells se$a Incidence (Cumm)ParentUV-2237 2/5 5/5 382Clone46 0/5 0/5 0CIone38 0/5 1/5 0Clone42 0/5 4/5 0CIonel5 0/5 3/5 16Clone18 0/5 5/5 95Clone3l 0/5 5/5 437CIone44 1/5 5/5 389Clone43 1/5 5/5 465Clone 9 1/5 5/5 616Clonel2 2/5 4/5 54Clone 22 2/5 5/5 237Clone30 2/5 5/5 272Clone 47 2/5 5/5 566Clone39 2/5 5/5 853Clone4l 3/5 4/5 10Clone 27 3/5 5/5 401CIone26 3/5 5/5 766Clone 5 3/5 5/5 824CIone33 4/5 5/5 132Clone 25 4/5 5/5 132Clone34 4/5 5/5 245

    a C3H mice were given s.c. injections on the flank of 1 x 10'viable cells. The number of mice with metastaseswas determinedat time of death or 5 months after injection.

    b Mice without tumors were observed for 5 months.

    absolute correspondence among the three tests. UV-2237clone 12 produced the highest number of experimentallung metastases following i.v. injection but was only intermediate in its ability to metastasizefrom a s.c. site. Thus,clone 12 appears to be a clone that is adept at surviving inthe circulation to form lung metastasesbut maybedeficientin its invasive capabilities. Clone 12, however, appears tobe the exception and thus the significant degree of correspondence among the three tests for the other 20 clonespermits us to conclude that at least for this UV-2237 fibrosarcoma the convenient experimental metastasisassayapproximates rather well the more tedious measurementsofspontaneous metastasis.

    Summary

    In summary I wish to state that the heterogeneous natureof neoplasms with regard to a large number of characteristics has been well recognized. Populations of human andanimal neoplasms frequently demonstrate a great variationin chromosome number and DNA content (37, 54). Tumorsare known to be heterogeneous with regard to their antigenic properties (46), immunogenicity (29), hormone receptors (51), pigment production (25), metabolic characteristics (31), and growth rate (49), as well as in their susceptibility to a variety of cytotoxic drugs (23, 27). Not surprisinglythen, we have now demonstrated that neoplasms are alsoheterogeneouswith regard to invasion and metastasis,i.e.,that they contain a variety of subpopulations of cells withdiffering metastatic potentials.

    The development of a metastasis appears to be dependent on an interplay between host factors and intrinsiccharacteristics of the tumor cells. The processof metastasis

    HeterogenemTable

    414'of UV-2237fibrosarcomafor tumor growth andetastasis following i.v. injection ofcellsNo.of

    Time ofdeathNo.ofmicewith (days)

    mice with extrapulpulmo-monarySource

    ofcellsnary

    me- metastatastases sest' MedianRangebParent

    223719/19 17/19 2118—26Clone4l3/50/5 15283-152Clone460/51/5 15257-152CIone472/51/5 15255-152Clone384/51/5 15250-152Clonel53/51/5 8636-152CIone424/51/5 6342-152Clone

    125/5 2/5 4237-73Clone185/5 3/5 5739-91Clone55/5 3/5 3634-127Clone

    315/5 3/5 2727Clone305/5 4/5 4937-81Clone335/5 4/5 4525-82Clone445/5 4/5 3734—44Clone265/5 4/5 2420-24Clone275/5 5/5 3833-38Clone435/5 5/5 3624-58Clone345/5 5/5 3624-36Clone395/5 5/5 3434-92Clone255/5 5/5 3428-36Clbne95/5 5/5 2727-36Clone

    224/5 5/5 24 22-34of 1 x 10' viablecells.

    I. J. Fidler

    a C3H mice were given i.v. injectionsMetastaseswere determined at autopsy.

    b Surviving mice were killed and autopsied on Day 152 afterinjection.

    is highly selective, and the metastatic lesion represents theend point of several destructive events that only a few cellscan survive.Only a few cells within a primary neoplasmmayactually invade blood vessels, and of these even fewer willsurvive transport, can attach firmly to the endothelium ofsmall blood vessels,will undergo extravasation, will evadehost defenses, and will grow into tumor foci.

    We may conclude the following: (a) the outcome ofmetastasis is dependent to a large extent on a selectionprocess that favors the survival and growth of a specialsubpopulation of cells. Therefore, studies that compareproperties of tumor cells obtained from primary tumors andthose obtained from their metastases may be helpful towardthe identification of some of the properties of metastasticcells; (b) the possible existence of highly metastatic variantcells within a primary tumor suggeststhat we no longer canconsider a neoplasmto be a uniform entity. Indeed, effortsto design effective therapeutic agents and proceduresshould be directed toward the few but fatal metastaticsubpopulations. Therapeutic efforts that are directedagainst all neoplastic cells without regard to their biologicalbehavior in vivo may be unproductive; (C) tumor variantswith differing metastatic potentials selected in a variety oftumor systems could be a useful tool for answering questions regarding the biology of metastasis and in particularfor testing new therapeutic approaches to cancer.

    Acknowledgments

    This presentation would not have been possible were itnot for the contributions of numerous investigators in thefield of metastasis. In particular, I wish to acknowledge my

    2656 CANCER RESEARCH VOL. 38

    on May 31, 2021. © 1978 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • Tumor Heterogeneity and Metastases

    teacher and friend, Dr. Irving Zeidman, for introducing meto and guiding me in the field of the biology of cancermetastasis.

    References1. Baserga, R., and Saffiotti, U. Experimental Studies on Histogenesis of

    Blood-BorneMetastases.Arch. Pathol.,59: 26-34,1965.2. Brunson, K. E., Beaftie, G., and Nicolson, G. L. Selection and Altered

    Properties of Brain-Colonizing Metastatic Melanoma. Nature, 272: 543-545, 1978.

    3. Chew, E. C., Josephson, R. L., and Wallace, A. C. Morphologic Aspectsof theArrestof CirculatingCancerCells.In: L. Weiss(ad.),FundamentalAspects of Metastasis, pp. 121-150. Amsterdam: North Holland PublishingCo., 1976.

    4. Cliffton, E. E., and Agostino, D. Factors Affecting the Development ofMetastaticCancer.Effectof Alterationsin Clotting Mechanism.Cancer,15: 276-283, 1962.

    5. Chiffon, E. C., and Grossi, C. E. The Rationale of Anticoagulants in theTreatmentof Cancer.J. Med.,5: 107-113,1974.

    6. Cloudman, A. M. Organophilic Tendencies of Two Transplantable Tumorsof the Mouse.CancerRae.,7: 585-591, 1947.

    7. del Regato, J. A. Pathways of Metastatic Spread of Malignant Tumors.SeminarsOncol.,4: 33—38,1977.

    8. Dorland, W. A. Dorland's Illustrated Medical Dictionary, Ed. 24. Philadelphia: W. B. Saunders Co., 1965.

    9. Dunn,T. B. Normaland PathologicAnatomyof the ReticularTissueinLaboratoryMice,with a Classificationand Discussionof Neoplasms.J.Natl. Cancer Inst., 14: 1281-1333, 1954.

    10. Ewing, J. Neoplastic Diseases,Ed. 3, Chap. 4. Philadelphia:W. B.SaundersCo., 1928.

    11. Fidler, I. J. Metastasis:QuantitativeAnalysisof Distributionand FateofTumor Emboli Labeled with “I-5-lodo-2'.deoxyuridine. J. NatI. CancerInst., 45: 733—782,1970.

    12. Fidler, I. J. The Relationship of Embolic Homogeneity, Number, Size andviability to the Incidence of Experimental Metastasis. European J.Cancer,9: 223-227,1973.

    13. Fidler, I. J. Selection of Successive Tumor Lines for Metastasis. NatureNew Biol., 242: 148-149, 1973.

    14. Fidler, I. J. Immune Stimulation-Inhibition of Experimental Cancer Metastasis.CancerAss.,34: 491-498,1974.

    15. Fidler, I. J. Mechanisms of Cancer Invasion and Metastasis. In: F. F.Baker (ed), Cancer:A ComprehensiveTreatise,Vol. 4, pp. 101-131.New York: Plenum Press, 1975.

    16. Fidler, I. J., and Krlpke, M. L. Metastasis Results from PreexistingVariantCellswithin a MalignantTumor.Science,197:893-895,1977.

    17. Fidler, I. J., and Nicolson, F. L. Organ Selectivity for Survival and Growthof B16 Melanoma Variant Tumor Lines. J. NatI. Cancer Inst., 57: 1199-1202, 1976.

    18. Fisher, B., and Fisher, E. R. The Interrelationship of Hematogenous andLymphatic Tumor Cell Dissemination. Surg. Gynecol. Obstet., 122: 791-798, 1966.

    19. Fisher, B., and Fisher, E. R. The Organ Distribution of Disseminated“Cr-labeledTumor Cells. Cancer Rca., 27: 412-420, 1967.

    20. Fisher, E. R., and Fisher, B. RecentObservationson the ConceptofMetastasis.Arch. Pathol.,83:321-324,1967.

    21. Folkman, J. Tumor Angiogenesis. In: F. F. Baker (ad.), Cancer: AComprehenisve Treatise, Vol. 3, pp. 355-388. New York: Plenum Press,1975.

    22. Franks, L. M. Structure and Biological Malignancy of Tumors. In: S.Garattini and G. Franchi (ads.), Chemotherapy of Cancer DisseminationandMetastasis,pp. 71-78.NewYork: RavenPress,1973.

    23. Fugi, H., and Mihich, E. Selection for High Immunogenicity in DrugResistant Sublines of Murine LymphomasDemonstratedby PlaqueAssay. Cancer Res., 35: 946-952, 1975.

    24. Gasic, G. J., Gasic, T. B., Galanti, N., Johnson, T., and Murphy, S.Platelet-TumorCell Interaction in Mice. The Role of Plateletsin theSpreadof MalignantDisease.Intern.J. Cancer,11:704—718,1973.

    25. Gray, J. M., and Pierce, G. B. Relationship between Growth Rate andDifferentiation of Melanomaln Vivo. J. NatI. Cancerlnst., 32: 1201-1211,1964.

    26. Greene,H. S. N., and Harvey,E. K. The RelationshipbetweentheDissemination of Tumor Cells and the Distribution of Metastases. CancerRes., 24: 799-811, 1964.

    27. Hakannson,L., and Troupe, C. On the Presencewithin Tumors ofClones That Differ in sensitivity to Cytostatic Drugs. Acta Pathol.Microbiol.Scand.A,82:32-40,1974.

    28. Hilgard,P.TheRoleof BloodPlateletsin ExperimentalMetastases.Brit.J. Cancer,28:429-436,1973.

    29. Killion, J. J., and Kollmorgen, G. M. Isolation of Immunogenic TumorCellsbyAffinity Chromatography.Nature,259:674-676,1976.

    30. Kinsey, D. L. An Experimental Study of Preferential Metastasis. Cancer,13: 674-676, 1960.

    31. Kirccuta, I., Mustea, I., Rogozaw, I., and Simu, G. Relations betweenTumor and Metastases.I. Aspectsof the CrabtreeEffect. Cancer,18:978-984, 1965.

    32. Krlpke,M. L. Latency,HistologyandAntigenicityof TumorsInducedbyUftraviolet Light in Three Inbred Mouse Strains. Cancer Roe., 37: 1395-1400, 1977.

    33. Kripke, M. L., Fidler, I. J., and Gruys, E. Heterogeneity of MetastaticPotential in Cells. Proc. Am. Assoc. Cancer Res.. 19: 213, 1978.

    34. LiII, P. H., and Kripke, M. L. Growth Patterns of Ultraviolet Light-InducedFibrosarcomas in Subcutaneous, Peritoneal, and Vascular Compartments of Syngeneic Recipients. Transplantation, 25: 86-87, 1978.

    35. Liotta, L. A., Kleinerman,J., and Saidel, G. M. The Significance ofHematogenousTumor Cell Clumps in the-MetastaticProcess.CancerRes.,36: 889-894,1976.

    36. Luria, S. E., and DelbrOck, M. Mutations of Bacteria from Virus SensitivIty to virus Resistance. Genetics, 28: 491-51 1, 1943.

    37. Makino, 5. The Chromosome Cytology of the Ascites Tumor of Rats,with SpecialReferenceto the Conceptof the StemlineCell. Intern.Rev.Cytol., 6: 26-84, 1957.

    38. Nicolson, G. L., and Brunson, K. w. Organ Specificity of Malignant 816Melanomas:In vivo Selection for Organ Preferenceof Blood-BorneMetastasis.GannMonographCancerRae.,20: 15-24,1977.

    39. Nicoison,G.L.,andWinkelhake,J. L.OrganSpecificityof Blood-BorneTumorMetastasisDeterminedby CellAdhesion?Nature,255:230-232,1975.

    40. Nowell, P. C. The Clonal Evolution of Tumor Cell Populations. Science,194: 23-28. 1976.

    41. Ozaki, T., Yoshida, K., Ushijima, K., and Hayashi, H. Studies on theMechanismsof Invasion in Cancer. II. In vivo Effects of a FactorChemotacticforCancerCells.Intern.J.Cancer,7:93-100,1971.

    42. Paget,S.TheDistributionofSecondaryGrowthinCanceroftheBreast.Lancet, 1: 571-573, 1889.

    43. Parks, R. C. Organ-Specific Metastasis of a Transplantable ReticulumCellSarcoma.J. NatI.CancerInst.,52:971-973,1974.

    44. PIlgrim, H. I. The Kinetics of the Organ Specific Metastasis of aTransplantable Reticuloendothelial Tumor. Cancer Res., 29: 1200-1205,1969.

    45. Potter, M., Fahey, J. L., and Pilgrim, H. I. Abnormal Serum Protein andBone Destruction in Transplantable Mouse Plasma Cell Neoplasm (Multiple Myeloma). Proc. Soc., Exptl. Biol. Med., 94: 327-333, 1957.

    46. Prehn, R. T. Analysis of Antigenic Heterogeneity within Individual 3-Methylcholanthrene-InducedMouseSarcomas.J. NatI.CancerInst.,45:1039-1045,1970.

    47. Salsbury,A. J. TheSignificanceof the CirculatingCancerCell. CancerTreat.Rev.,2: 55-72,1975.

    48. Sato, H., and Suzuki, M. Deformability and Viability of Tumor Cells byTranscapillary Passage, with Reference to Organ Affinity of Metastasisin Cancer. In: L. Weiss (ed.), Fundamental Aspects of Metastasis, pp.311-317.Amsterdam:NorthHollandPublishingCo., 1976.

    49. Schabel,F. M., Jr. Conceptsfor SystemicTreatmentof Micrometastases. Cancer, 35: 15-24, 1975.

    50. Siegel, 5. Nonparametric Statistics for the Behavioral Sciences. NewYork:McGraw-HillBookCo., 1956.

    51. Sluyser, M., and VanNie, R. Estrogen Receptor Content and Hormoneresponsive Growth of Mouse Mammary Tumors. Cancer Res., 34: 3253-3257,1974.

    52. Sugarbaker,E. V., Cohen,A. M., and Ketcham,A. S. Do MetastasesMetastasize? Ann. Surg., 174: 161-166, 1971.

    53. Sugarbaker, E. V., and Ketcham, A. 5. MechanIsms and Prevention ofCancer Dissemination: An Overview. Seminars Oncol., 4: 19-32, 1977.

    54. SuzukI, N., Frapart, N., Grdina, D. J., Meistnch, N. L., and Withers, H.R.CellCycleDependencyof Metutatic LungColonyFormation.CancerRes.,37:3690-3693,1977.

    55. Warren, B. A. Environment ofthe Blood-Borne Tumor Embolus Adherentto Vessel Wall. J. Med., 4: 150-177, 1973.

    56. Weiss, L. The Cell Periphery, Metastasis and Other Contact Phenomena.Frontiers Biol., 7:289-332,1967.

    57. Weiss,L. BiophysicalAspectsof the MetastaticCascade.In: L. Weiss(ad.), Fundamental Aspects of Metastasis, pp. 51-70. Amsterdam: NorthHollandPublishingCo., 1976.

    58. WeIss, L. A Pathobiologic Overview of Metastasis. SemInars Oncol., 4:5-17, 1977.

    59. Willis, R. A. The Spread of Tumors In the Human Body. London:Butterworths, 1972.

    60. Wood, S., Jr. Experimental Studies of the Intravascular Dissemination ofAscitlc V2 Carcinoma Cells in the Rabbit. with Special Reference toFibrinogen and Fibrinolytic Agents. Bull Schweiz. Akad. Med. Wise., 20:92-121, 1964.

    61. Zeldman,I. Metastasis:A Reviewof RecentAdvances.CancerRes.,17:157-162, 1957.

    62. Zeidman,I. The Fateof CirculatingTumorCells.I. Passageof Callsthrough Capillaries. Cancer Res., 21: 38-39, 1961.

    63. Zeldman, I., and Buss, J. M. Transpulmonary Passage of Tumor CallEmboli.CancerRes.,12:731-733,1952.

    2657SEPTEMBER1978

    on May 31, 2021. © 1978 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • ic

    Fig. 1. Anin vivo assayto determinethe metastaticpotentialof a transplantableneoplasm.B16melanomacellsare injecteds.c. into theexternalearof aC57BL/6mouse.Threeweekslaterwhenthe tumor is established(a),the ear is amputated.Six weeksthereafter,metastasesin the draining regionallymphnodeaswell as in the lungsareevident (b). Thecomponentsof this assayare shown in C.

    2658 CANCER RESEARCHVOL. 38

    on May 31, 2021. © 1978 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • Tumor Heterogeneity and Metastases

    ‘@@

    .‘

    P1hZ,

    p

    2d

    -1a@@

    .@@ @â€Ĩb @‘4@

    Fig. 2. Theformationof anexperimentalpulmonarymetastasis.B16melanomacellswereinjectediv. into syngeneicmice.Theinjectedmicewerekilledat different time points thereafter, and their lungs were fixed and serially sectioned. Tumor cells (arrows, a to i) can be identified by their melanin content. a,immediate (initial) arrest of tumor cells in the microvasculature; b, 1 hr postinjection; C, 4 hr postinjection; d, 1 day postinjection; e, 2 days postinjection(note cell division); f, 4 days postinjection; g, 6 days postinjection; h, 9 days postinjection; I, 12 days postinjection;j, 18 days postinjection. a to i, x 450;j,x 100. H & E.

    I

    ,

    F

    2f.E@

    SEPTEMBER 1978 2659

    1@@

    ‘l@.

    (f'....JII1p S,@ .1@

    .@

    i@@r

    on May 31, 2021. © 1978 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • I. J. Fidler

    a

    @2I

    2660 CANCERRESEARCHVOL. 38

    on May 31, 2021. © 1978 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • 1978;38:2651-2660. Cancer Res Isaiah J. Fidler MetastasisTumor Heterogeneity and the Biology of Cancer Invasion and

    Updated version

    http://cancerres.aacrjournals.org/content/38/9/2651

    Access the most recent version of this article at:

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected] at

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://cancerres.aacrjournals.org/content/38/9/2651To request permission to re-use all or part of this article, use this link

    on May 31, 2021. © 1978 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/content/38/9/2651http://cancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://cancerres.aacrjournals.org/content/38/9/2651http://cancerres.aacrjournals.org/


Recommended