Sie sind auf Seite 1von 12

THYROID

Volume 8, Number 8, 1998


Mary Ann Liebert, Inc.

Anaplastic Thyroid Carcinoma:


Behavior, Biology, and Therapeutic Approaches
KENNETH B. AIN

ABSTRACT

Anaplastic thyroid carcinoma (ATC), although exceedingly rare, is the most aggressive solid tumor known. Early
studies on the effects of different therapies may be biased by the inclusion of responsive "small cell" ATC pa¬
tients, which are now known to be mostly lymphoma patients. Local control of disease with surgery and/or ex¬
ternal beam radiotherapy (XRT) is of fundamental importance to enhance survival. Ultimately, nearly all ATC
patients die from their disease, which is widely metastatic. Development of effective systemic chemotherapy agents
would provide the best chance for long-term survival of patients. Early preliminary data suggest that paclitaxel
may be helpful, although no agent has yet been identified to result in dramatic improvements in survival. Select
patients may benefit from aggressive multimodal therapy, although it is important to provide appropriate pallia¬
tive care when desired.

INTRODUCTION There are regional differences as well as changes over


time. In recent reports, ATC cases constituted 7.5% of thy¬
certainly the
is roid cancer cases in Tel Aviv (4), 4.7% in Bombay (5), 5%
Anaplastic
mans.
aggressive and lethal solid
most
It is
fortunate that
(ATC)
thyroid carcinoma

ATC is
the least
tumor afflicting hu¬
common of thy¬
in a national Italian survey, and 1% in Arsizio, Italy (6).
There is a distinct temporal trend of reduction in the pro¬
roid neoplasms; however, this has also made it difficult to portion of thyroid carcinomas found to be ATC. In Bom¬
study and test therapeutic strategies. Spindle and giant cell bay, the relative proportion of ATC declined from 7.7%
ATC remains the neoplasm most recalcitrant to known in 1969-1973 to 4.2% in 1989-1993, at the same time
therapeutic modalities and present the greatest challenge that the total number of thyroid carcinomas increased by
for clinical management. The rarity of this cancer, its fail¬ 3.5-fold (5). Likewise, between f979 and 1989, the pro¬
ure to respond to treatment, and the relative lack of active portion of ATC declined from 11% to 5% in Italy as a
research regarding its management, justifies its classifica¬ whole and from 10% to 1% in Arsizio, Italy (6). Similar
tion as a true "orphan disease." decreases in the frequency of ATC have been ascribed to
improvements in socioeconomic status (7). Potential addi¬
tional explanations for this decline include enhanced
EPIDEMIOLOGY pathologic discrimination with reclassification of some tu¬
mors as medullary cancers or lymphomas and earlier treat¬
The incidence of thyroid carcinomas in the United States ment of differentiated carcinomas to prevent anaplastic
is approximately 17,200 cases per year (1). Although some transformation.
older studies described up to 20% of thyroid cancers as Female patients typically outnumber male patients by a
ATC or "undifferentiated" (2), the most recent results from ratio of from 3.1 to 1.2 to 1 in various series (3-5,8-10).
the Surveillance, Epidemiology and End Results (SEER) The mean age at diagnosis is from 57 to 67 years with a
Program show 251 ATC cases of 15,698 thyroid cancers range of ages from 15 to 91 years (3,5,9-11). This is def¬
(1973-1991), indicating that ATC constitutes 1.6% of thy¬ initely a disease of older adults with 90% over the age of
roid cancers in the United States (3) for an estimated an¬ 50 years in one series (4) and only 4 patients under the age
nual incidence of less than 300 cases. of 40 years in another series of 121 ATC patients (10).

Thyroid Nodule and Oncology Clinical Service, Division of Endocrinology and Molecular Medicine, Department of Internal Medicine,
University of Kentucky Medical Center, Thyroid Clinic, Veterans Affairs Medical Center Lexington, Kentucky.
The author is the recipient of NCI Grant CA58935 and support from the Ephraim McDowell Cancer Research Foundation (Lexington,
KY).
715
716 AIN

PRESENTATION ity to interpret the effectiveness of different therapeutic in¬


terventions.
The majority of ATC patients present with a rapidly Mean reported survival times of ATC patients range
growing thyroid mass (4,10,11). Nearly half of the patients from 2.5 to 7.4 months (6,8,10,23), while median survival
have symptoms of hoarseness and dyspnea, with some not¬ is reported as 4 to 12 months (9,11,18). In a representa¬
ing dysphagia or cervical pain (4,9,11). Clinicians should tive series, 21% of patients died within the immediate post¬
also be alert to unusual presentations such as ball-valve operative period (8). From 82% to 90% of patients expire
type respiratory obstruction (12,13), gastrointestinal symp¬ by 6 months (8,23). In the largest American series of ATC
toms from métastases (14,15), or superior vena cava syn¬ patients, those presenting with clinical evidence of only lo¬
drome (16,17). From 28% to 71% of patients have a his¬ cal disease had an 8.1-month median survival compared
tory of previous or concurrent differentiated thyroid to a 3.3-month median survival of patients presenting with
carcinomas, either follicular or papillary (and its variants) distant métastases (p < 0.001) (10). Likewise, evidence of
(9,10,18). It appears likely that more extensive histopatho- distant disease at presentation results in a mortality risk
logic analysis reveals higher rates of associated differenti¬ ratio of 3.2 (95% confidence limits at 2.0-5.1) compared
ated thyroid carcinoma. with patients with only localized disease (3). The rare ATC
Physical examination typically demonstrates palpable patients with apparent long-term survival are typically
thyroid masses that are usually multiple and bilateral. younger, have small primary foci of localized ATC, and
Nearly 80% of patients have tumors greater than 5 cm in have been aggressively treated with surgical resection, ex¬
diameter at presentation (11). Cervical adenopathy is pres¬ ternal radiation therapy, and usually systemic chemother-
ent in more than 40% of patients and vocal cord paraly¬ apy (9,10,23).
sis in around 30% (4,11,17). At least 50% of patients have
distinct métastases at presentation, of whom around 80%
have métastases in the lung, 15% in bone, and 13% in PATHOLOGY AND IMMUNOHISTOCHEMISTRY
the brain (4,10). This probably underestimates the predilec¬
tion for aggressive distant spread since an autopsy study In its gross appearance, ATC is frequently seen as a tan-
of 15 ATC patients revealed all to have pulmonary metas- white, fleshy, large thyroid tumor that infiltrates extrathy-
tases, 80% with bone métastases, 60% to soft tissue of the roidal tissues and has regions of necrosis and hemorrhage
neck, 27% to trachea, and 53% with other soft tissue (24). Three predominant histological patterns are com¬
métastases (2). monly seen, often coexisting: spindle cell in 53%, giant cell
in 50%, and squamoid in f 9% (8,22). Common features
to all of these patterns are large foci of necrosis, marked
CLINICAL COURSE AND MORTALITY invasiveness, and predilection for growth into the muscu¬
lar walls of medium-sized veins and arteries (angiotropism)
ATC has a relentless and deadly clinical course with (22). Cytological assessment reveals anaplasia with bizarre
rapid progression and dissemination. Clinicians may ob¬ tumor cells and a high mitotic rate (24). Up to 10% of
serve tumors double in volume over a period of several ATC tumors have osteoclast-like giant cells, which exhibit
days to a week. With rare exception, nearly all patients die immunohistochemical staining for vimentin and KP-1 an¬
from their tumor within several months. This outcome is tibody (thought to be specific for histiocytic cells), sug¬
essentially unaltered by any known therapies, although (as gesting that these cells may be nonneoplastic in origin and
will be discussed below) mortality may be delayed. It is may have resulted from a coalescence of histiocytic
difficult to rely on some published literature for valid mor¬ mononuclear cells (22,25,26). As would be expected for
tality information because of the failure to discriminate this aggressive tumor, the rate of proliferation exceeds that
"small cell" ATC (representing lymphomas, medullary can¬ of any other type of primary thyroid malignancy, while the
cers, or insular carcinomas with slower growth rates or rate of apoptosis is the lowest (27), accounting for its ex¬
better responses to treatment) from spindle and giant cell ceedingly rapid rate of growth.
ATC. For example, Rossi et al. (19) compiled survival data As introduced earlier, current pathological opinion sug¬
on 96 cases of "ATC" (most treated with external radio¬ gests that the former classification of small cell ATC is a
therapy), comprised of 51 giant cell ATC patients with misnomer and does not represent any true subset of ATC.
long-term (5-year) survival of 4 patients (8%), and 45 pa¬ In a rigorous analysis, Wolf et al. (28) studied 68 cases of
tients with "small cell" ATC whose long-term survival ex¬ putative small cell ATC with a variety of immunohisto¬
ceeded 28%. Likewise, the 1969 report by Rafia (20) chemical markers for B-cells and T-cells, as well as leuko¬
claims a 24% long-term (approximately 5-year) survival of cyte common antigen, vimentin, thyroglobulin, calcitonin,
ATC patients, in contrast to a contemporary report of 53 cytokeratins, carcinoembryonic antigen, and chromo-
ATC patients with 92% mortality before 1 year (21), which granin. Sixty-seven cases were found to be lymphomas (63
could be explained by a 25% subset of "small cell" ATC cell, 2 of indeterminate phenotype, and 2 large cell) and
cases. In another study at the Mayo Clinic, only 3 of 82 the remaining case was morphologically similar to small
(3.6%) ATC patients were reported to have a reported 5- cell lung carcinoma. In another series of 31 unselected ATC
year survival (11). A re-analysis of the histology of these cases, similar immunohistochemical reclassification
3 patients by Rosai et al. (22) revealed them to consist of demonstrated 7 cases to be lymphomas, which included all
2 cases of lymphoma and 1 case of medullary carcinoma, 3 of the "small-cell" type and 4 of 16 putative giant cell
leaving the Mayo Clinic series with no long-term ATC sur¬ ATC cases (29). Corrected nosology of the "small cell"
vivors. This nosological problem may also affect our abil¬ ATC designation reveals lymphomas, insular carcinomas,
ANAPLASTIC THYROID CARCINOMA 717

and medullary carcinomas, all of which have better prog¬ duction of thyroid hormone with clinical thyrotoxicosis
noses than ATC. In general, a series of ATC patients with due to an activating mutation of the thyrotropin receptor
long-term survivors should be suspected of having lym¬ (47).
phomas, medullary carcinomas, or insular carcinomas mis-
classified as ATC (8,22,24,30).

Immunohistochemistry of ATC shows varied results in GENESIS OF ANAPLASTIC CARCINOMA


different series (8,10,26,31,32). Generally, positive stain¬
ing for thyroglobulin is seen in from none to 55% of cases. It appears likely that ATC represents a terminal dedif¬
Vimentin is positive in 23% to 94%, keratins or cytoker- ferentiation of pre-existing differentiated carcinoma of the
atin positive in 12% to 80%, epithelial membrane antigen thyroid follicular cell in most, if not all, cases. This is based
positive in 8% to 55%, ai-antichymotrypsin positive in on indirect, although convincing, evidence. A large portion
48%, coexpression of vimentin and keratin in 39-75%, of ATC cases develop in longstanding goiters or in the con¬
desmin negative in all cases, and 30% of cases may be neg¬ text of pre-existing, incompletely treated papillary or fol¬
ative for all of the above markers. Squamoid variants of licular thyroid cancers. Likewise, careful examination of
ATC typically stain for carcinoembryonic antigen (8). primary ATC tumors reveals coexisting areas of differen¬
Cases staining positive for calcitonin are medullary can¬ tiated thyroid carcinoma in 80% to 90% of cases (17,21).
cers, those positive for factor VHI-associated antigen are Such differentiated tumors include papillary and follicular
angiosarcomas or hemangioendotheliomas (32,33), and varieties, as well as Hiirthle-cell variants (48). Some pathol-
those positive for smooth muscle actin or muscle-specific ogists describe areas of histological transition from differ¬
actin are usually leiomyosarcomas (34,35). Immunohisto¬ entiated carcinoma to ATC and occasional tiny foci of ATC
chemistry has proven a powerful tool to discriminate tu¬ may be found within well-differentiated carcinomas (4,24).
mors with distinct clinical behaviors and to suggest differ¬ Two studies evaluated metaplastic transition into ATC
ent therapeutic approaches. in fatal thyroid cancers. Harada et al. (49) autopsied 27
There are a few variants of ATC with distinctive fea¬ consecutive cases of fatal thyroid carcinomas and found
tures. A recent report suggests that just under 6% of ATC 10 cases of uniform ATC, 4 cases of uniform well-differ¬
cases constitute the paucicellular variant of ATC, charac¬ entiated carcinoma, and 9 cases with both tumor types co¬
terized by rapid growth of an infiltrating fibrotic thyroid existing, suggesting a natural progression of well-differen¬
mass presenting with compressive symptoms and rapid tiated carcinomas to ATC. Furthermore, 26% of 86
death. Histopathology reveals large areas of fibrosis, foci patients who died of papillary carcinoma demonstrated
of calcification, regions of infarction, atypical spindle cells spindle and giant cell metaplasia in their tumors, but only
obliterating large blood vessels and immunohistochemical 2% of 241 papillary carcinoma survivors had such meta¬
staining for carcinoembryonic antigen, actin, and cytoker- plasia in their primary malignancies, usually as minute foci,
atin (36). These cases could have easily been confused for if present. Once large amounts of metaplasia became evi¬
Riedel's struma. Other unusual ATC cases have apprecia¬ dent, the average survival shortened to 5 months (50).
ble areas of osteosarcomatous differentiation (carcinosar- A contrary view of Wallin et al. (51) derived from their
coma). These areas stain for vimentin, but not keratin, and analysis of 11 of 17 ATC tumors with coexistent differ¬
have prominent osteoid formation and focal calcification, entiated carcinoma. Utilizing cytophotometric DNA analy¬
suggesting a mixed epithelial-mesenchymal differentiation sis, they found that in only 36% of the cases the differen¬
(37). In two reports of this type of ATC, recurrent and tiated carcinoma foci shared DNA aneuploidy with the
metastatic tumor foci were purely osteosarcomatous and ATC component, suggesting that the majority of ATC cases
were both fatal at 26 months, although similar cases in the arise de novo rather than through clonai transformation
literature generally died within 5 months (38,39). of differentiated carcinomas. However, Galera-Davidson
Pure squamous cell carcinoma of the thyroid is extremely et al. (52) evaluated 8 similar cases and found DNA ane¬
rare, although metaplastic squamoid elements are not un¬ uploidy in both the differentiated and ATC components.
common components of ATC (40). The tumor is highly in¬ Their conclusion was that differentiated thyroid carcino¬
vasive and usually far advanced at time of presentation. mas with aneuploidy represented cases with a higher risk
Most patients expire within 6 months, although Simpson of ATC transformation. In fact, most ATC cases are found
and Carruthers (41) noted two patients alive after 4 years. to be aneuploid (53) and analysis of ATC DNA content
Insular carcinoma has previously been considered a va¬ has little, if any, prognostic significance (54-56). It is likely
riety of ATC and has also been called a solid variant of that aneuploidy, as a feature of the anaplastic phenotype,
follicular carcinoma or a poorly differentiated type of pap¬ may be a later metaplastic development, rather than a pre¬
illary carcinoma (22). This cancer is composed of nests of requisite for anaplastic transformation.
uniform cells and small follicles in an "insular" architec¬ An early observation of primary cultures of ATC re¬
tural pattern. The cells range from uniformly round to pleo- vealed that rounded epithelial cells spontaneously trans¬
morphic with nuclear features typical of papillary cancer formed into spindle and giant cell forms, recapitulating the
(intranuclear inclusion, grooves, and folds), rare mitoses, morphology of the original tumor (57). This is likely to be
and scant fine cytoplasm (42). Insular carcinomas have a an epiphenomenon of culture conditions because I see such

prognosis falling between ATC and differentiated carcino¬ reversible phenotypic changes in most thyroid carcinoma
mas. They may represent as much as 4% to 7% of thyroid cell lines, of all varieties of tumor histology, consequent to
cancers (43). Unlike ATC, insular carcinomas frequently nutritional status or degree of confluency. In contrast, a
retain iodide uptake and respond to radioiodine therapy fascinating report focuses attention on a more likely fac¬
(44^16). A unique case demonstrated autonomous pro¬ tor involved in transformation of thyroid carcinomas to
718 AIN

ATC. A case of well-differentiated follicular carcinoma permits complete thyroid resection, this did not alter out¬
contained multiple tiny foci of ATC, which uniquely im- come in a small series of these patients (73). In some cases,
munostained for p53 protein overexpression consequent to FNA produces insufficient diagnostic material, necessitat¬
mutation of this gene. When microdissected, these specific ing open biopsy.
foci were shown to share the identical mutation in exon 6 In most cases, features of rapid tumor enlargement and
exhibited by the lung and brain métastases of ATC that invasive growth are reliable clues to the presence of ATC.
killed this patient 8 years later (58). Other investigators In other cases, a single tiny focus of ATC in an otherwise
have found a high incidence of p53 mutations in ATC, but well-differentiated thyroid carcinoma primary tumor or
rare, if any such mutations in coexisting differentiated can¬ metastasis is revealed after resection of the differentiated
cer foci, suggesting that inactivating mutations of this tu¬ tumor. Because the latter cases may have improved sur¬
mor-suppressor gene are important in triggering progres¬ vival (17) and be amenable to therapy, it is important to
sion from differentiated to anaplastic carcinomas (59-62). carefully review the pathological findings of all thyroid tu¬
This issue is not completely clear because some researchers mors.
have found similar p53 mutations or immunohistochem¬ Nuclear and radiographie imaging procedures have lit¬
istry in well-differentiated thyroid carcinomas (63,64). tle role in the diagnosis of ATC, but may be helpful in stag¬
Other oncogenes that appear to be highly expressed in ATC ing disease. Astute clinicians refrain from using intravenous
include c-myc (65) and Nm23 (65,66). Activation of ras contrast containing iodine for radiographie studies of thy¬
oncogenes is implicated in the development of ATC from roid tumors because this would compromise radioiodine
the study of Stringer et al. (67) in which DNA, extracted treatment of differentiated carcinomas. Once the diagno¬
from four ATC tumors, was transfected into murine 3T3 sis of ATC is established, however, there is no reason to
cells, resulting in malignant transformation and the ex¬ avoid such studies. Nuclear scans using radioiodine trac¬
pression of human -ras. In another experiment, an ATC ers are usually not appropriate because ATC does not con¬
cell line, TTA-1, was the recipient of normal human chro¬ centrate iodine and optimal scanning preparation requires
mosome 11 using microcell-fusion-mediated chromosome induction of hypothyroidism, which further weakens de¬
transfer (68). This caused suppression of tumorigenicity bilitated patients. Sometimes radioiodine scanning may
and growth in soft-agar, suggesting that ATC cells were demonstrate the extent of coexistent differentiated thyroid
deficient in activity of another putative tumor suppressor cancer; however, differentiated cancers do not contribute
gene located on chromosome 11. much to the mortality of ATC patients. Thallium-201 nu¬
Considering the fatal consequences of ATC, demonstra¬ clear scans may assist in discerning ATC métastases that
tion of its genesis from differentiated thyroid carcinomas evade detection by computerized tomographic radio-
places a particular burden on the clinician to aggressively graphic scanning or magnetic resonance imaging. Consid¬
eliminate differentiated cancers in their earliest stages, be¬ ering the rapid growth of this tumor type, métastases rarely
fore anaplastic transformation can take place. At the very remain undetected for long.
least, reductions in tumor cell numbers may lessen the
chance for oncogene mutations that could result in ATC.
SURGICAL MANAGEMENT

DIAGNOSTIC APPROACH Primary surgical management of ATC is an uncertain


and controversial topic. In view of the dismal prognosis,
Fine-needle aspiration (FNA) biopsy has supplanted there is a distinct tendency to severely limit tumor and thy¬
most other methods of diagnosing thyroid malignancies, roid resection so as to merely provide confirmatory histo¬
both to avoid unnecessary resection of benign lesions and logical samples and protect the airway. Frequently, this is
to optimize the therapeutic approach by discriminating the not a matter of choice because grossly invasive disease of¬
type of malignancy. This can be accomplished without ten precludes significant resection. It appears that a total
morbidity as an office procedure. FNA is an appropriate thyroidectomy is justifiable if cervical and mediastinal dis¬
diagnostic modality for the evaluation of ATC presenting ease can be resected with reasonable morbidity, particu¬
as a thyroid or cervical mass. In a series of 113 cases of larly to avoid upper airway obstruction (74). Patients with
ATC evaluated by a FNA biopsy (69), 89.7% were cor¬ more complete thyroid resections seem to have longer sur¬

rectly identified with ATC, whereas 5% were thought to vival (75). Junor et al. (76) evaluated 91 ATC patients
be another type of thyroid malignancy. Just under 3% of treated between 1961 and 1986. The 5% of patients with
the FNA biopsies were inadequate for diagnosis and an total thyroidectomies, along with the 31% of patients with
equal number were rated suboptimal. Reasons for inade¬ partial thyroidectomies, had significantly prolonged sur¬
quate FNA results included: tumor regressive changes vival compared with patients who were merely biopsied.
(necrosis, hemorrhage, and leukocytic infiltration), exten¬ Kobayashi et al. (77) found that more complete tumor re¬
sive fibrosis, and sampling errors in regions of differenti¬ section prolonged the mean survival from 2 months to 6
ated carcinoma. Others have also found that coexistent ar¬ months. Although a study of 82 ATC patients by Nel et
eas of differentiated thyroid carcinoma contributed to FNA al. (11) was compromised by the inclusion of 19 patients
sampling error (70). FNA biopsies may reveal the presence with probably lymphoma (ie, "small cell" ATC), there was
of osteoclast-like giant cells and provide sufficient cellular a distinct trend toward prolonged survival with more com¬
material for immunocytochemical analysis (71,72). Unfor¬ plete resections. The increased likelihood of small primary
tunately, although early detection of ATC by FNA biopsy tumors to receive the most aggressive resections, introduces
allows diagnosis while the primary tumor size is small and obvious selection bias to account for this finding. Hermann
ANAPLASTIC THYROID CARCINOMA 719

et al. (78) found that reoperation of patients after tumor response after completion of the full 6-week course and
recurrence to provide only palliative tracheostomy
a was 68% retained local disease control until their death. Al¬
associated with a mean survival of 64 days compared with though all patients died of distant métastases and all de¬
a mean survival of 4f 2 days in those patients successfully veloped moderate pharyngoesophagitis and tracheitis from
treated with a complete resection of the recurrent tumor the XRT, the median survival of 12 months was quite long.
after a primary surgery that involved radical tumor resec¬ This method appears to provide the best opportunity for
tion. local disease control in the absence of complete surgical re¬
Prophylactic tracheostomies may not provide much help section; however, there was no control group to test the
to patients with unresected local disease. In 32 ATC pa¬ value of the chemoradiosensitization and local morbidity
tients treated with tracheostomies (of which 45% were pro¬ was significant. A later review of 32 patients treated with

phylactic against later respiratory complications), survival a different schema of hyperfractionated local XRT (initial
was significantly lower (2 months compared with 5 dose of 5 Gy followed by 1 Gy given 4 times daily, with
months) than for 45 patients who did not receive tra¬ 3 hours between doses, for 5 days each week to a total
cheostomies. Many of the patients with tracheostomies had dose of 30-45 Gy) resulted in a median survival of less
local wound healing complications that were considered to than 6 months and showed no benefit from concomitant
prevent postoperative radiation therapy (79). doxorubicin administered to half of the patients at 40
mg/m2 every 3 weeks (83). Skin and esophageal toxicities
appeared more severe than conventional XRT and 2 pa¬
RADIOTHERAPY tients developed radiation myelopathy despite the use of
posterior spinal cord shielding at 30 to 40 Gy, no expo¬
Local control of disease is an important component of sure to doxorubicin, and low total spinal doses of 39.9 and
clinical management. External beam radiotherapy (XRT) 48.3 Gy, respectively. Because the latency of myelopathy
can provide substantial benefits in this regard, although is around 20 months, far longer than these patients sur¬
ATC should be considered as radioresistant relative to vived, it is possible that the risk for this complication could
other solid malignancies. In one series, analysis of survival be greater than observed. In an earlier report from the same
in ATC patients demonstrated a prolonged mean survival institution, Simpson (84) emphasized that toxkity with this
of 6 months in patients receiving local XRT of more than variety of hyperfractionated XRT was quite severe.
30 Gy as compared with 2 months in patients receiving Tennvall et al. (85,86) treated 33 ATC patients with
lower XRT doses (77). Similarly, a retrospective assess¬ chemosensitized XRT before and after attempted surgical
ment of 51 ATC patients treated between 1970 and 1986, resection of local disease. They used two regimens of hy¬
with 22 patients having distant métastases at the time of perfractionated XRT, 1 Gy or 1.5 Gy fractions (given twice
diagnosis and an additional 21 patients developing distant daily for 5 days each week to totals of 30 Gy before surgery
métastases after diagnosis, noted that only 1 such patient and 16 Gy after surgery), each given with concomitant
had a successful total thyroidectomy without gross resid¬ doxorubicin at 20 mg administered 1 to 2 hours prior to
ual local tumor (80). Among those patients, cumulative each week's first XRT fraction. Median survival was sim¬
XRT doses less than 30 Gy were associated with a median ilar with each regimen (3.5 and 4.5 months, respectively);
survival of 0.6 months with no patients alive at 1 year, however, 4 of the 33 patients were alive without evidence
while cumulative XRT doses greater than or equal to 30 of disease at the time of publication, for periods ranging
Gy were associated with a median survival of 3.3 months from 30 to 84 months, and 48% of the patients achieved
with a 10% survival at 1 year. The subgroup of patients local disease control. With that approach, the preoperative
who completed their course of XRT without evidence of hyperfractionated XRT may have improved the resectabil-
residual local disease, despite distant métastases, had a 7.5- ity of the primary tumors. Hyperfractionation generally
month median survival compared with a 1.6 month me¬ permits the administration of higher total radiation doses
dian survival for patients with residual local ATC tumor. with less toxicity; however, the addition of chemosensiti-
Ultimately, all of the patients in this series died of ATC. zation is still unsettled in reference to types of agents, doses,
Both of these studies demonstrate the importance of local and acceptable toxicity. Nonetheless, for patients with
disease control in prolonging survival and the utility of residual local postoperative disease, there is sufficient evi¬
XRT to provide this control, particularly when complete dence to support the use of XRT (in some form, provid¬
surgical resection is not possible. On the other hand, Junor ing at least 30 Gy) to provide a degree of local disease con¬
et al. (76) noted that, despite an 80% total response rate trol and prolongation of survival, even in the presence of
to local XRT and 39% with complete responses, there was lethal distant métastases.
no apparent enhancement of survival by XRT in their large

patient cohort.
In 1983, Kim and Leeper (81) reported the use of hy¬ SYSTEMIC CHEMOTHERAPY
perfractionated XRT, augmented with low-dose doxoru¬
bicin (10 mg/m2) as a radiosensitizer, for local control of With one-half of ATC patients presenting with distant
ATC. Their final report in 1987 (82) revealed the results disease, the rest developing distant métastases at some time
of this treatment in f 9 patients with ATC. Patients received after presentation, and even patients with local control of
5760 cGy XRT over 6 weeks administered in 160 cGy disease dying of distant spread of tumor, successful sys¬
doses twice daily (4 hours apart) for 3 consecutive days of temic therapy is the fundamental key to survival. Unfor¬
each week with doxorubicin administered once weekly, 1.5 tunately, there has not yet been any chemotherapeutic
hours before XRT. There was 84% complete local tumor agent or combination of agents with sufficient antineo-
720 AIN

plastic activity against ATC to alter its fatal outcome. How¬ tients was reported using a combination of bleomycin, cy¬
ever, in rare patients with partial or complete responses, clophosphamide, and 5-fluorouracil (100).
delay in the progression of disease may significantly pro¬ Consideration of the relative ineffectiveness of antineo¬
long lives. As in many studies of ATC, because lymphomas plastic agents in ATC suggests an active role for one or
constitute a majority of the "small cell" ATC subtype, usu¬ more cellular mechanisms associated with chemotherapy
ally with greater response to chemotherapy than giant and resistance. Satake et al. (101) summarized their results, in¬
spindle cell ATC, some positive responses may be spurious. cluding two previous studies, investigating the expression
For ATC, doxorubicin (Adriamycin®, Pharmacia, of P-glycoprotein (MDR1 gene product), MRP (multidrug
Dublin, OH) has been used most commonly, but mono¬ resistance-associated protein, MRP1 gene product), LRP
therapy with this drug has been quite disappointing. A re¬ (major vault protein), and mutations of DNA topoiso-
view of chemotherapy for advanced thyroid carcinoma by merase Il-alpha (TOPII) in 10 ATC cell lines and 3 pri¬
Poster et al. (87) found most studies to have patients with mary tumors. Using immunohistochemistry for these pro¬
only a few partial responses (PR) to this agent, and little teins, reverse-transcriptase polymerase chain reaction
evidence of any complete responses (CR). Likewise, in 16 (RT-PCR) analysis for respective mRNAs, as well as DNA
ATC patients treated with multiple chemotherapy drugs, sequence analysis to assess for TOPII mutations, they
as single agents or in different combinations, Gottlieb and found all cell lines to express MRP (protein and mRNA),
Hill (88) found that doxorubicin provided only one PR, 7 positive for LRP proteins (8 positive by RT-PCR), and
with the same investigators later reporting only 2 PRs in 6 expressing MDR1 (protein and mRNA). All cell lines and
9 ATC patients treated with doxorubicin monotherapy tissues expressed TOPII and there were no mutations in
(89). One decade later, Shimaoka et al. (90) verified the this gene. All 3 of the tumors were positive for MRP and
failure of doxorubicin monotherapy, documenting only LRP, but MDR1 was seen in only 1 tumor. Most or all of
one PR in 2f patients receiving this modality, despite hav¬ the cell lines were resistant to doxorubicin, vincristine, cis¬
ing previously reported a summary of the literature in 1980 platin, and etoposide, with 2 resistant to bleomycin. P-gly¬
showing 23% PR to this agent alone (91). Reviewing many coprotein and MRP are membrane proteins that pump
of these same studies in 1987, Ahuja and Ernst (92) re¬ chemotherapy agents out of cells, resulting in resistance to
ported a similar 22.1% total response rate for 77 ATC pa¬ those agents. MRP has a similar spectrum of drug trans¬
tients. Other chemotherapy agents have been even less ef¬ port activity as p-glycoprotein, except that it is less able to
fective as monotherapy, although minimal activity has been transport paclitaxel (Taxol®, Bristol-Myers Squibb Oncol¬
claimed for bleomycin (87), etoposide (VP-16) (93), cis¬ ogy, Princeton, NJ) or colchicine (102). LRP has similar
platin (93), and methotrexate (23). drug transport properties in the membrane of intracellular
Chemotherapy combinations for ATC have typically in¬ vesicles, resulting in sequestration and ultimate exocytosis
cluded doxorubicin. Two studies evaluated doxorubicin in of chemotherapy agents, while mutations in TOPII may al¬
combination with cisplatin. The best results were reported ter this potential drug target (103). Our laboratory has re¬
by Shimaoka et al. (90) who noted 3 CRs and 3 PRs in 19 ported roughly similar results as above, except that all of
ATC patients for a 33% response rate. However, Williams our 6 ATC cell lines expressed MRP1 mRNA by RT-PCR
et al. (94) terminated their trial after finding only 1 PR in and none expressed MDR1 mRNA (104). Besides serving
a small number of patients, claiming the treatment to be as an explanation for the failure of systemic antineoplas¬
toxic and ineffective. These results are similar to the dis¬ tic therapies, these results suggest that MRP and LRP may
mal findings reported in a recent series of 14 ATC patients provide appropriate targets for inactivation, in order to re¬
(95). Addition of bleomycin to this regimen (ie, doxoru¬ store clinical response to standard chemotherapies.
bicin, cisplatin, and bleomycin) seemed to increase the re¬
sponse rate in a subset of 5 ATC patients out of a larger
series, resulting in 2 CRs and 1 PR, as well as a long mean COMBINED MODALITY TREATMENT
survival of 16 months (range 3-58 months) (96). Sokal and
Harmer (97) substituted vincristine for cisplatin (ie, doxo¬ Given the rapid, locally invasive growth of the ATC pri¬
rubicin, vincristine, and bleomycin) and found 4 PRs in a mary tumor, as well as its propensity for widespread dis¬
subset of 5 ATC patients from a larger group of advanced tant metastasis, with distant disease already evident at the
thyroid cancer, noting that the mean survival was only 7.6 time of diagnosis, it is logical to combine XRT with sys¬
months despite this response rate. Melphalan was added temic chemotherapy. Clearly, clinical studies have demon¬
to this regimen (ie, doxorubicin, vincristine, bleomycin, strated that local tumor control gained with XRT can pro¬
and melphalan) by Bukowski et al. (98) resulting in 2 PRs long survival in the face of unresected local disease, while
out of 5 giant cell ATC patients, with no noticeable dif¬ concomitant chemotherapy can interact to radiosensitize
ference in survival between responders and nonresponders. the primary tumor and may also enhance the ability of the
There are two promising studies of chemotherapy com¬ surgeon to clear otherwise unresectable gross disease. The
binations that do not contain doxorubicin. Kober et al. weak link in this therapeutic schema is the lack of a dis¬
(99) used a combination of cisplatin, vincristine, and mi- tinctively effective systemic antineoplastic regimen.
toxantrone in f 5 ATC patients, and reported 4 CRs and Nonetheless, several clinical studies have suggested the pos¬
6 PRs resulting in prolonged mean survival of 15.5 months sibility of enhanced survival and are discussed below.
and median survival of 20.8 months, compared with 3.8 One of the largest cohorts of ATC patients managed with
months and 4.5 months, respectively, in nonresponders. multimodality therapy consisted of 71 patients seen at one
Another high response rate of 7 responders of 9 ATC pa¬ institution between 1973 and 1989 (105). Despite preop-
ANAPLASTIC THYROID CARCINOMA 721

erative combinations of chemotherapy agents and XRT, These studies are remarkable in that, although there was
only 13% of patients were able to undergo successful com¬ little effect on general mortality, each regimen was associ¬
plete resection of their primary tumors. Nearly three-quar¬ ated with a small number of unique patients with long-
ters of these patients had distant métastases at presenta¬ term survival.
tion and only 9% of the cohort survived for more than a Two case reports delineate particularly aggressive com¬
year, suggesting the inadequacy of the systemic component bined modality therapies with apparently good results.
of this treatment regimen. Likewise, Schlumberger et al. Casterline et al. (110) reported one ATC patient treated
(106) treated 20 ATC patients with either doxorubicin and with a total thyroidectomy and unilateral radical neck dis¬
cisplatin (60 mg/m2 and 90 mg/m2 every 4 weeks) or mi- section, followed by XRT (60 Gy over 40 days) with acti-
toxantrone (14 mg/m2 every 4 weeks), both regimens in nomycin-D therapy started during the last week of irradi¬
combination with XRT (17.5 Gy in 7 fractions between ation (0.5 mg weekly for 10 doses). A later course of
days 10-20 of the first 4 chemotherapy cycles), and noted actinomycin-D therapy (total dose of 26.5 mg) was sup¬
only 1 patient alive with each treatment regimen (at 34 plemented with immunotherapy utilizing a methanol-ex-
and 40 months, respectively) and all patients suffering se¬ tractable residue of BCG for 12 monthly courses. This pa¬
vere pharyngoesophagitis and tracheitis. Although the lo¬ tient was reported as free of clinical disease at the final
cal response appeared good, the systemic response was 48-month follow-up. A recent report of Sweeney et al.
poor, and both long-term survivors were from among the (Ill) described a 61-year-old woman presenting with mas¬
few patients with successful complete local surgical resec¬ sive and unresectable local disease treated with a combi¬
tions. nation of: hydroxyurea (f g twice daily for 11 doses), 5-
Tallroth et al. (107) divided 47 patients into 1 of 4 dif¬ fluorouracil (800 mg/m2 per day continuously over 5 days),
ferent combinations of chemotherapy and XRT. The first paclitaxel (20 mg/m2 continuously over 5 days), and XRT
group received methotrexate with XRT (30—40 Gy); the (2 Gy daily for 5 days). This combination was adminis¬
second group received bleomycin, cyclophosphamide, and tered every 2 weeks for 7 cycles (total XRT dose of 70
5-fluorouracil with XRT; the third group received the same Gy). Although there was severe mucositis, dysphagia, and
treatment as the second group, with the addition of surgi¬ weight loss requiring total parenteral nutrition, the primary
cal resection in midcourse and XRT increased in dose to tumor regressed sufficiently to perform a near-total thy¬
46 Gy and hyperfractionated; the fourth group was treated roidectomy and unilateral modified neck dissection at 6
similarly to the third group except that doxorubicin was months from the initiation of therapy. Pathological evalu¬
the only chemotherapy. The majority of patients in each ation revealed necrotic tissue with no evidence of viable
treatment group demonstrated some response to treatment tumor. Despite the transient appearance of pulmonary nod¬
with median and mean survivals (months) of: 9 and 9 for ules on computerized axial tomography (CAT) scanning,
group 1; 4 and 15 for group 2; 4 and 15 for group 3; and the patient has remained free of evidence of ATC for at
4 and 5 months for group 4. There was 1 long-term sur¬ least 38 months (E.E. Vokes, personal communication). A
vivor of more than f 2 years in group 2; 2 such survivors key component of both of these cases is the absence of dis¬
(3.5 and fl years) in group 3; and 1 survivor for more cernible distant disease at the time of initiating aggressive
than 10 months in group 4. Although there did not seem chemoradiotherapy.
to be a clear advantage to any particular treatment regi¬
men, the long-term survival of over 8% appears more fa¬
vorable than otherwise expected. BIOLOGICAL INVESTIGATIONS AND FUTURE
In a recent Japanese series of 37 ATC patients CLINICAL DIRECTIONS
(1971-1993) treated with different chemotherapy agents
in combination with XRT, the median survival was in¬ In the face of an aggressive, rare, and usually untreat-
creased to 8 months as compared with an unspecified group able cancer, further research is essential. Prospective clin¬
receiving palliative treatment with 2-month median sur¬ ical trials have no reasonable chance of acquiring enough
vival. In a similar result as reported in the other series subjects to evaluate more than a few likely therapeutic ap¬
above, there were 3 unusual long-term survivors for more proaches. In addition, there is no clear evidence that cur¬
than 3 years. This is also similar to the result seen with 19 rently available therapies are likely to provide sufficient ac¬
ATC patients treated with bleomycin, cyclophosphamide, tivity against this disease. ATC cell lines furnish the
and 5-fluorouracil in combination with hyperfractionated investigator a versatile model system to study the cellular
XRT to a dose of 30 Gy, followed by thyroidectomy and and molecular biology particular to this malignancy, as
an additional XRT to 15 Gy (108). The 9 patients pre¬ well as to devise and test novel antineoplastic strategies. In
senting with distant disease or locally invasive tumor had the past decade, the number of ATC cell lines have in¬
a median survival of 7 months, while the 10 patients with creased dramatically. These cells can be grown in culture
local, noninvasive disease had a median survival of 12 dishes or as xenograft tumors in athymic mice and appear
months and included 3 long-term survivors (at 31, 61, and to appropriately reproduce many of the features of the pri¬
80 months, respectively). Twelve of the 19 patients were mary tumors, including cytokinetics (112), radioresistance
able to undergo thyroidectomy consequent to the preop¬ (113), invasive, and metastatic behavior (114). Cell line
erative combined therapy. A similar approach with 5 ATC monolayers provide an opportunity for inexpensive and
patients used preoperative doxorubicin and cisplatin with rapid screening of potential antineoplastic agents; however,
postoperative XRT, resulting in an average survival of 11.2 active agents so identified must be tested against xenograft
months and 1 long-term survivor at 31 months (109). tumors to verify preclinical effectiveness. For example,
722 AIN

suramin appears to be a potent cytotoxic agent in ATC except for partial sensitivity to doxorubicin and cisplatin
monolayer cultures, yet this agent accelerates the growth in only 1 cell line. Xenografts of primary ATC tumors in
of the same cell lines grown as xenograft tumors (115). nude mice showed only slight inhibition by doxorubicin
Analysis of cellular receptors for hormones and growth treatment and no enhancement of this effect with cy¬
factors may predict responses to their ligands. BHT-101 clophosphamide coadministration (131). We have investi¬
cells display estrogen and progesterone receptors and are gated chemosensitivity with a panel of six distinct ATC cell
inhibited by tamoxifen (116). The absence of high-affinity lines, grown in monolayer cultures as well as in nude mice
binding sites for thyrotropin on ATC cell membranes, de¬ as xenografts, in an attempt to define new active agents.

spite an intact adenylate cyclase transduction system, ac¬ Paclitaxel exhibited unique antineoplastic activity against
curately correlates with the absence of clinical ATC tumor all of the cell lines as monolayers and xenograft tumors
responses to endogenous or exogenous thyrotropin (117). were dramatically diminished with complete disappearance

Transforming growth factor-/? (TGF-/3) seems to function of some of them (132). Consideration of MRP as a mech¬
as an endogenous physiological inhibitor of thyroid follic¬ anism for ATC chemotherapy resistance, with its decreased
ular cell growth and differentiation, suggesting that the low activity against intracellular paclitaxel accumulation, sug¬
expression of its receptor, seen in ATC, may contribute to gests a reason for paclitaxel's activity in this disease. Con¬
its rapid growth and undifferentiated status (118). Al¬ sequently, an ongoing phase 2 human trial of paclitaxel
though the expression of platelet-derived growth factor monotherapy for ATC has demonstrated notable clinical
(PDGF) receptor B-type has been thought to be restricted response rates, with 1 complete response and 6 partial re¬
to mesenchymal and glial cells, the ATC cell line, C643, sponses in the first 11 évaluable patients, for a 64% re¬
expresses this receptor in a form that has more complex sponse rate (unpublished results). These preliminary results
glycosylation than in normal human fibroblasts (119). suggest this agent to be one of the most active for
ATC cells have been found to express angiogenesis factors chemotherapy of this disease.
(120), IGF-I-like growth-stimulating factors (121), and Despite the absence of radioiodine uptake in ATC tu¬
highly glycosylated forms of CD97 with unknown func¬ mors and lack of expression of sodium-iodide symporter
tion (122). We have shown that ATC cell lines express so- in ATC cell lines (our unpublished data), there may still be
matostatin receptor subtypes 1, 3, and 5 (123). Because so- a role for nuclear medicine therapy of ATC. Astatine-211
matostatin receptor agonists can inhibit cellular is an -emitting (mean energy 6.8 MeV) halogen isotope
proliferation (124), this provides a rationale for evaluat¬ with a half-life of 7.2 hours. In two separate studies
ing receptor subtype-specific somatostatin analogues for (133,134) human ATC expiants were grown as subcuta¬
antineoplastic activity. neous xenografts in nude mice. They were given a single
ATC can be associated with aseptic purulent effusions, intravenous or intraperitoneal injection of 370 or 740 kBq
leukocytosis, and fever. Iwasa et al. (125) described such of sodium astatine-211. By some unknown mechanism,
a case in which the tumor demonstrated immunohisto¬ ATC was able to concentrate astatine-211 2.6- to 12.5-
chemical staining for granulocyte-macrophage colony- fold over iodine-125 (134), taking 11% to 28% of the ad¬
stimulating factor (GM-CSF). In a similar patient, culture ministered dose per gram of tumor tissue in 6 of 8 ATC
of ATC cells from a malignant pleural effusion yielded the expiants (133). In normal thyroid glands, astatine uptake
KHM-5M cell line, which expressed interleukin 8 (IL-8) is blocked by C104~, TcO-t", and SCN~, much like iodide;
and GM-CSF, producing neutrophil infiltrations in nude however, propylthiouracil and thiouracil treatment in¬
mouse xenografts (126). The K-119 cell line was estab¬ creases astatine uptake, contrary to the effect on iodide up¬
lished from another ATC patient with marked neutrophilia take (133), implying a different route for astatine concen¬
and also spontaneously secreted GM-CSF, with augmented tration in ATC tissue. There are several impediments to
secretion in the presence of tumor necrosis factor (TNF), research with astatine-211, including the need to be in close
IL-la, and IL-1/3 (127). Evaluation of a panel of ATC cell proximity to a sufficiently powerful cyclotron to generate
lines revealed that secretion of GM-CSF spontaneously, or the isotope, the unknown chemical toxicity of astatine, and
after stimulation by IL-la, TNF-a, or phorbol esters, is the difficulty of dealing with radiotoxicity of an -emitter
common (128). The role of such immunoactive factors is to normal tissues. Nonetheless, this is a provocative idea
not well understood; however, Baba et al. (129) reported for potential new therapies.
an interesting case of a woman with ATC (the source of
the ATC cell line, KOA-2) who received intratumoral in¬
jections of -432/fibrinogen solution in a futile attempt CURRENT SUGGESTIONS FOR MANAGEMENT
at immunotherapy. The patient'slymphocytes were fused
with a human lymphoblastoid cell line (HO-323), pro¬ The current therapeutic armamentarium for ATC is very
ducing hybridoma that secreted monoclonal IgM (3C5) limited, but can be life-prolonging, or at least palliative, if
a
and reacted with cell membranes of KOA-2, as well as a appropriately utilized. Rare individuals with tiny foci of
number of other ATC tumors and some normal tissues. anaplastic carcinoma may enjoy long-term survival with
This suggests the possibility of future application of im¬ aggressive therapy. Certainly, any opportunity to enroll
munotherapy for this disease. ATC patients into clinical therapy trials should be taken,
A few investigators have attempted to define activity of because their scarcity impedes the progress of prospective
chemotherapeutic agents using ATC cell lines in monolayer clinical research and delays the development of new treat¬
culture. Asakawa et al. (130) used 3 ATC cell lines to eval¬ ments. These suggestions on management reflect one ap¬
uate the response to doxorubicin, cisplatin, etoposide, and proach to care and cannot be considered definitive in this
carboplatin. These cell lines were resistant to those agents changing field.
ANAPLASTIC THYROID CARCINOMA 723

Careful management of the local tumor mass can extend tic thyroid carcinoma: A study of 70 cases. Am J Clin Pathol
survival. If resectable, this should be the first aspect of treat¬ 83(2):135-158.
ment, provided that vital structures are not severely com¬ 9. Demeter JG, De Jong SA, Lawrence AM, Paloyan E 1991
promised. This is helpful, even in the presence of known Anaplastic thyroid carcinoma: Risk factors and outcome.
distant métastases, provided that distinct disease is not clin¬ Surgery 110(6):956-963.
10. Venkatesh YS, Ordonez NG, Schultz PN, Hickey RC,
ically more advanced than the local tumor. Complete re¬ Goepfert H, Samaan NA 1990 Anaplastic carcinoma of the
section of gross disease is frequently impossible and local
hyperfractionated external beam radiotherapy is distinctly thyroid: A clinicopathologic study of 121 cases. Cancer
66(2):321-330.
helpful in this circumstance. It is uncertain whether 11. Nel CJC, van Heerden JA, Goellner JR, Gharib H, Mc-
chemoradiosensitization provides sufficient therapeutic
benefit to justify increased radiation morbidity, but this may Conahey WM, Taylor WF, Grant CS 1985 Anaplastic car¬
cinoma of the thyroid: A clinicopathologic study of 82 cases.
be considered. Likewise, it is unclear whether prophylactic Mayo Clin Proc 60(l):51-58.
tracheostomies enhance patient survival or comfort. 12. Carter N, Milroy CM 1996 Thyroid carcinomaca using fa¬
Systemic therapy is usually necessary, if aggressive treat¬ tal laryngeal occlusion. J Laryngol Otol 110:1176-1178.
ment is desired, because most patients either present with 13. Lee WC, Walsh RM 1996 Anaplastic thyroid carcinoma pre¬
distant disease or develop these métastases soon after di¬ senting pharyngeal mass with ball-valve type obstruc¬
as a
agnosis. Although traditionally, most patients have re¬ tion of thelarynx. J Laryngol Otol 110:1078-1080.
ceived chemotherapy centered on doxorubicin, initial data 14. Lip GYH, Jaap AJ, McCruden DC 1992 A presentation of
on paclitaxel suggests that this agent has at least as much anaplastic carcinoma of the thyroid with symptomatic in-
activity, if not more, than doxorubicin or other agents. tra-abdominal métastases. Br J Clin Pract 46(2):143-144.

Certainly, effective systemic therapies are the cornerstone 15. Phillips DL, Benner KG, Keeffe EB, Traweek ST 1987 Iso¬
to radically improving the dismal survival data. lated metastasis to small bowel from anaplastic carcinoma.
Limited reported experience with combination modali¬ J Clin Gastroenterol 9(5):563-567.
ties suggests that aggressive and appropriate combinations 16. Glikson M, Feigin RD, Libson E, Rubinow A 1990 Anaplas¬
of surgery, radiation, and chemotherapy may provide some tic thyroid carcinoma in a retrosternal goiter presenting as
benefit. Preoperative chemotherapy and radiation may en¬ fever of unknown origin. Am J Med 88:81-82..
hance the completion of a surgical resection of the primary 17. Aldinger KA, Samaan NA, Ibanez M, Hill CS Jr 1978
tumor. Enhanced networking of clinical investigators may Anaplastic carcinoma of the thyroid: A review of 84 cases
of spindle and giant cell carcinoma of the thyroid. Cancer
identify sufficient subjects to obtain prospective data on 41(6):2267-2275.
the value of combination modality therapy for ATC.
Finally, because the typical ATC patient is far-advanced
18. Spires JR, Schwartz MR, Miller RH 1988 Anaplastic thy¬
roid carcinoma: Association with differentiated thyroid can¬
in years, often with other comorbidities, the option of pal¬
cer. Arch Otolaryngol Head Neck Surg 114(1 ):40-44.
liative care with or without local XRT must be considered. 19. Rossi R, Cady B, Meissner WA, Sedgwick CE, Werber J
It is essential to provide patients with appropriate comfort 1978 Prognosis of undifferentiated carcinoma and lym¬
measures and emotional support.
phoma of the thyroid. Am J Surg 135:589-596.
20. Rafla S 1969 Anaplastic tumors of the thyroid. Cancer
23(3):668-677.
REFERENCES 21. Nishiyama RH, Dunn EL, Thompson NW 1972 Anaplastic
spindle-cell and giant-cell tumors of the thyroid gland. Can¬
1. Landis SH, Murray T, Bolden S, Wingo PA 1998 Cancer cer 30(1):113-127.
statistics, 1998. CA Cancer J Clin 48(l):6-29. 22. Rosai J, Saxén EA, Woolner L 1985 Session III: Undiffer¬
2. Thomas GG Jr, Buckwalter JA 1973 Poorly differentiated entiated and poorly differentiated carcinoma. Sem Diag
neoplasms of the thyroid gland. Ann Surg 177(5):632-642. Pathol 2(2):123-136.
3. Gilliland FD, Hunt WC, Morris DM, Key CR 1997 Prog¬ 23. Jereb B, Stjernswärd J, Löwhagen 1975 Anaplastic giant-
nostic factors for thyroid carcinoma: A population-based cell carcinoma of the thyroid: A study of treamtent and prog¬
study of 15,698 cases from the Surveillance, Epidemiology nosis. Cancer 35(5):1293-1295.
and End Results (SEER) Program 1973-1991. Cancer 24. LiVolsi VA 1990 Surgical Pathology of the Thyroid.
79(3):564-573. Philadelphia, W.B. Saunders Co., 1990:253-274.
4. Hadar , Mor C, Shvero J, Levy R, Segal 1993 Anaplas¬ 25. Gaffey MJ, Lack EE, Christ ML, Weiss LM 1991 Anaplas¬
tic carcinoma of the thyroid. Eur J Surg Oncol 19(6): tic thyroid carcinoma with osteoclast-like giant cells: a clin¬
511-516. icopathologic, immunohistochemical, and ultrastructural
5. Agrawal S, Rao RS, Parikh EM, Parikh HK, Borges AM, study. Am J Surg Pathol 15(2):160-168.
Sampat MB 1996 Histologie trends in thyroid cancer 26. Ordonez NG, El-Naggar AK, Hickey RC, Samaan NA 1991
1969-1993: A clinico-pathologic analysis of the relative pro¬ Anaplastic thyroid carcinoma: immunocytochemical study
portion of anaplastic carcinoma of the thyroid. J Surg On¬ of 32 cases. Am J Clin Pathol 96(l):15-24.
col 63:251-255. 27. Basólo F, Pollina L, Fontanini G, Fiore L, Pacini F, Baldanzi
6. Lampertico 1993 Anaplastic (sarcomatoid) carcinoma of A 1997 Apoptosis and proliferation in thyroid carcinoma:
the thyroid gland. Semin Diag Pathol 10(2):159-168. Correlation with bcl-2 and p53 protein expression. Br J Can¬
7. Bakiri F, Djemli FK, Mokrane LA, Djidel FK 1998 The rel¬ cer 75(4):537-541.
ative roles of endemic goiter and socioeconomic develop¬ 28. Wolf BC, Sheahan K, DeCoste D, Variakojis D, Alpern HD,
ment status in the prognosis of thyroid carcinoma. Cancer Haselow RE 1992 Immunohistochemical analysis of small
82(6):1146-1153. cell tumors of the thyroid gland: An Eastern Cooperative
8. Carcangiu ML, Steeper T, Zampi G, Rosai J 1985 Anaplas¬ Oncology Group study. Hum Pathol 23(11):1252-1261.
724 AIN

29. Hölting T, Möller , Tschahargane C, Meybier H, Buhr H, Fatal thyroid carcinoma: Anaplastic transformation of ade-
Herfarth C 1990 Immunohistochemical reclassification of nocarcinoma. Cancer 39(6):2588-2596.
anaplastic carcinoma reveals small and giant cell lymphoma. 50. Hutter RVP, Tollefsen HR, de Cosse JJ, Foote FW Jr, Frazell
World J Surg 14(3):291-295. EL 1965 Spindle and giant cell metaplasia in papillary car¬
30. Nusynowitz ML 1991 Differentiating anaplastic thyroid car¬ cinoma of the thyroid. Am J Surg 110:660-668.
cinomas. J Nucl Med 32(7):1363-1364. 51. Wallin G, Bäckdahl M, Tallroth-Ekman E, Lundell G, Auer
31. LiVolsi VA, Brooks JJ, Arendash-Durand 1987 Anaplas¬ G, Löwhagen 1989 Co-existent anaplastic and well dif¬
tic thyroid tumors: Immunohistology. Am J Clin Pathol ferentiated thyroid carcinomas: A nuclear DNA study. Eur
87:434-442. J Surg Oncol 15(l):43-48.
32. Shvero J, Gal R, Avidor I, Hadar , Kessler E 1988 Anaplas¬ 52. Galera-Davidson , Bibbo M, Dytch HE, González-Cám-
tic thyroid carcinoma: A clinical, histologie, and immuno¬ pora R, Fernández A, Wied GL 1987 Nuclear DNA in
histochemical study. Cancer 62(2):319-325. anaplastic thyroid carcinoma with a differentiated compo¬
33. Lamovec J, Zidar A, Zidanik 1994 Epithelioid angiosar- nent. Histopathology 11:715-722.
of the thyroid gland: Report of two cases. Arch Pathol
coma 53. Mark J, Ekedahl C, Dahlenfors R, Westermark1987 Cy-
Lab Med 118:642-646. togenetical observationsin five human anaplastic thyroid
34. Kawahara E, Nakanishi I, Terahata S, Ikegaki S 1988 carcinomas. Hereditas 107:163-174.
Leiomyosarcoma of the thyroid gland: A case report with a 54. Ekman ET, Wallin G, Bäckdahl M, Löwhagen T, Auer
comparative study of five anaplastic carcinomas. Cancer 1989 Nuclear DNA content in anaplastic giant-cell thyroid
62(12):2558-2563. carcinoma. Am J Clin Oncol 12(5):442-446.
35. Thompson LDR, Wenig BM, Adair CF, Shmookler BM, 55. Klemi PJ, Joensuu H, Eerola E 1988 DNA aneuploidy in
Heffess CS 1997 Primary smooth muscle tumors of the thy¬ anaplastic carcinoma of the thyroid gland. Am J Clin Pathol
roid gland. Cancer 79(3):579-587. 89(2):154-159.
36. Wan S-K, Chan JKC, Tang S-K 1996 Paucicellular variant 56. Yoshida A, Asaga T, Masuzawa C, Kawahara S, Yanoma
of anaplastic thyroid carcinoma: A mimic of Riedel's thy¬ S, Shimizu A, Harada M 1993 The growth activity and DNA
roiditis. Am J Clin Pathol 105(4):388-393. ploidy in anaplastic carcinoma transplanted to nude mice.
37. Blasius S, Edel G, Grünert J, Böcker W, Schmid KW 1994 Nippon Geka Gakkai Zabshi 94(2):177-181.
Anaplastic thyroid carcinoma with osteosarcomatous dif¬ 57. Fisher ER, Gregorio R, Shoemaker R, Horvat B, Hubay C
ferentiation. Pathol Res Pract 190:507-510. 1974 The derivation of so-called "giant-cell" and "spindle-
38. Donnell CA, Pollock WJ, Sybers WA 1987 Thyroid carci- cell" undifferentiated thyroidal neoplasms. Am J Clin Pathol
nosarcoma. Arch Pathol Lab Med 111:1169-1172. 61(5):680-695.
39. Pascal-Vigneron V, Schneegans O, Weryha G, Floquet J, 58. Matias-Guiu X, Villanueva A, Cuatrecasas M, Capella G,
Olivier P, Mayot D, LeClere J 1993 Osteogenic anaplastic De Leiva A, Prat J 1996 p53 in a thyroid follicular carci¬
carcinoma of the thyroid. Thyroid 3(4):319-323. noma with foci of poorly differentiated and anaplastic car¬
40. Harada T, Shimaoka K, Katagiri M, Shimizu M, Hosoda Y, cinoma. Pathol Res Pract 192:1242-1249.
Ito 1994 Rarity of squamous cell carcinoma of the thy¬ 59. Dobashi Y, Sugimura H, Sakamoto A, Mernyei M, Mori M,
roid: Autopsy review. World J Surg 18(4):542-546. Oyama T, Machinami R 1994 Stepwise participation of p53
41. Simpson WJ, Carruthers J 1988 Squamous cell carcinoma gene mutation during dedifferentiation of human thyroid
of the thyroid gland. Am J Surg 156:44^6. carcinomas. Diagn Mol Pathol 3(1):9-14.
42. Zakowski MF, Schlesinger K, Mizrachi HH 1992 Cytologie 60. Fagin JA, Matsuo K, Karmakar A, Chen DL, Tang S-H,
features of poorly differentiated "insular" carcinoma of the Koeffler HP 1993 High prevalence of mutations of the p53
thyroid: A case report. Acta Cytol 36(4):523-526. gene in poorly differentiated human thyroid carcinomas. J
43. Pietribiasi F, Sapino A, Papotti M, Bussolati G 1990 Cyto¬ Clin Invest 91:179-184.
logie features of poorly differentiated 'insular' carcinoma of 61. Ito T, Seyama T, Mizuno T, Tsuyama N, Hayashi Y, Dohi
the thyroid, as revealed by fine-needle aspiration biopsy. Am , Nakamura N, Akiyama M 1993 Genetic alterations in
J Clin Pathol 94(6):687-692. thyroid tumor progression: association with p53 gene mu¬
44. Justin EP, Seabold JE, Robinson RA, Walker WP, Gurll NJ, tations. Jpn J Cancer Res 84:526-531.
Hawes DR 1991 Insular carcinoma: A distinct thyroid car¬ 62. Nakamura T, Yana I, Kobayashi T, Shin E, Karakawa K,
cinoma with associated iodine-131 localization. J Nucl Med Fujita S, Miya A, Mori T, Nishisho I, Takai S 1993 p53
32(7):1358-1363. gene mutations associated with anaplastic transformation of
45. Bal C, Padhy AK, Panda S, Kumar L, Basu AK 1993 "In¬ human thyroid carcinomas. Jpn J Cancer Res 83:1293-
sular" carcinoma of thyroid: A subset of anaplastic thyroid 1298.
malignancy with a less aggressive clinical course. Clin Nucl 63. Soares P, Cameselle-Teijeiro J, Sobrinho-Simöes M 1994 Im¬
Med 18(12):1056-1058. munohistochemical detection of p53 in differentiated,
46. Hassoun AAK, Hay ID, Goellner JR, Zimmerman D 1997 poorly differentiated and undifferentiated carcinomas of the
Insular thyroid carcinoma in adolescents: A potentially lethal thyroid. Histopath 24:205-210.
endocrine malignancy. Cancer 79(5):1044-1048. 64. Zou M, Shi Y, Farid NR 1993 p53 mutations in all stages
47. Russo D, Tumino S, Arturi F, Vigneri P, Grasso G, Pon- of thyroid carcinomas. J Clin Endocrinol Metab
tecorvi A, Filetti S, Belfiore A 1997 Detection of an acti¬ 77(4): 1054-1058.
vating mutation of the thyrotropin receptor in a case of an 65. Hoang-Vu C, Dralle H, Scheumann G, Maenhaut C, Horn
autonomously hyperfunctioning thyroid insular carcinoma. R, von zur Mühlen A, Brabant G 1992 Gene expression of
J Clin Endocrinol Metab 82(3):735-738. differentiation- and dedifferentiation markers in normal and
48. McDonald RJ, Wu S-Y, Jensen JL, Parker LN, Lyons KP, malignant human thyroid tissues. Exp Clin Endocrinol
Moran EM, Blahd WH 1991 Malignant transformation of 100:51-56.
a Hiirthle cell tumor: Case report and survey of the litera¬ 66. Zou M, Shi Y, Al-Sedairy S, Farid NR 1993 High levels of
ture. J Nucl Med 32(6):1266-1269. Nm23 gene expression in advanced stage of thyroid carci¬
49. Harada T, Ito , Shimaoka , Hosoda Y, Yakumaru 1977 nomas. Br J Cancer 68:385-388.
ANAPLASTIC THYROID CARCINOMA 725

67. Stringer BM, Rowson JM, Parkar MH, Seid JM, Hearn PR, Tibblin S 1994 Combined doxorubicin, hyperfractionated
Wynford-Thomas D, Ingemansson S, Woodhouse N, Goyns radiotherapy, and surgery in anaplastic thyroid carcinoma.
MH 1989 Detection of the -RAS oncogene in human thy¬ Cancer 74(4):1348-1354.
roid anaplastic carcinomas. Experientia 45:372-376. 87. Poster DS, Bruno S, Penta J, Pina , Catane R 1981 Cur¬
68. Yoshida A, Asaga T, Masuzawa C, Kawahara S 1994 Al¬ rent status of chemotherapy in the treatment of advanced
teration of tumorigenicity in undifferentiated thyroid carci¬ carcinoma of the thyroid gland. Cancer Clin Trials 4:
noma cells by introduction of normal chromosome 11. J 301-307.
Surg Oncol 55:170-174. 88. Gottlieb JA, Hill CS, Jr., Ibanez ML, Clark RL 1972
69. Us-Krasovec M, Golouh R, Auersperg M, Besic N, Rupar- Chemotherapy of thyroid cancer. An evaluation of experi¬
cic-Oblak L 1996 Anaplastic thyroid carcinoma in fine nee¬ ence with 37 patients. Cancer 30(3):848-853.
dle aspirates. Acta Cytol 40(5):953-958. 89. Gottlieb JA, Hill CS, Jr 1974 Chemotherapy of thyroid can¬
70. Bauman ME, Tao L-C 1995 Cytopathology of papillary car¬ cer with Adriamycin. Experience with 30 patients. Engl
cinoma of the thyroid with anaplastic transformation: A case J Med 290(4):193-197.
report. Acta Cytol 39(3):525-529. 90. Shimaoka K, Schoenfeld DA, DeWys WD, Creech RH, De-
71. Berry B, MacFarlane J, Chan 1990 Osteoclastomalike Conti R 1985 A randomized trial of doxorubicin versus
anaplastic carcinoma of the thyroid: Diagnosis by fine nee¬ doxorubicin plus cisplatin in patients with advanced thyroid
dle aspiration cytology. Acta Cytol 34(2):248-250. carcinoma. Cancer 56(9):2155-2160.
72. Lee JS, Lee MC, Park CS, Juhng SW 1996 Fine needle as¬ 91. Shimaoka 1980 Adjunctive management of thyroid can¬
piration cytology of anaplastic carcinoma with osteoclast- cer: Chemotherapy. J Surg Oncol 15:283-286.
like giant cells of the thyroid. Acta Cytol 40:1309-1312. 92. Ahuja S, Ernst 1987 Chemotherapy of thyroid carcinoma.
73. Pimpl W, Rieger R, Winkler J, Boeckl O, Galvan G 1990 J Endocrinol Invest 10:303-310.
Wert der präoperativen feinnadelpunktionszytologie in der 93. Hoskin PJ, Harmer C 1987 Chemotherapy for thyroid can¬
diagnose hochmaligner schilddrüsentumoren. Wiener Klin¬ cer. Radiother Oncol 10:187-194.
ische Wochenschrift 102(9):244-247. 94. Williams SD, Birch R, Einhorn LH 1986 Phase II evalua¬
74. Lore JM Jr 1991 Surgery for advanced thyroid malignancy. tion of doxorubicin plus cisplatin in advanced thyroid can¬
Otolaryngol Clin North Am 24(6):1295-1319. cer: A Southeastern Cancer Study Group trial. Cancer Treat
75. Tan RK, Finley RK III, Driscoll D, Bakamjian V, Hicks WL Rep 70(3):405^107.
Jr, Shedd DP 1995 Anaplastic carcinoma of the thyroid: A 95. Asakawa H, Kobayashi T, Komoike Y, Maruyama H,
24-year experience. Head Neck 17:41—48. Nakano Y, Tamaki Y, Matsuzawa Y, Monden M 1997
76. Junor EJ, Paul J, Reed NS 1992 Anaplastic thyroid carci¬ Chemosensitivity of anaplastic thyroid carcinoma and
noma: 91 patients treated by surgery and radiotherapy. Eur poorly differentiated thyroid carcinoma. Anticancer Res
J Surg Oncol 18:83-88. 17:2757-2762.
77. Kobayashi T, Asakawa H, Umeshita K, Takeda T, 96. De Besi P, Busnardo B, Toso S, Girelli ME, Nacamulli D,
Maruyama H, Matsuzuka F, Monden M 1996 Treatment Simioni N, Casara D, Zorat P, Fiorentino MV 1991 Com¬
of 37 patients with anaplastic carcinoma of the thyroid. bined chemotherapy with bleomycin, adriamycin, and plat¬
Head Neck 18:36-41. inum in advanced thyroid cancer. J Endocrinol Invest
78. Hermann M, Kober F, Hollinsky C, Heiss A 1990 Reein- 14:475-480.
griffe bei anaplastischen karzinomen und Sarkomen der 97. Sokal M, Harmer CL 1978 Chemotherapy for anaplastic
Schilddrüse. Wiener Klinische Wochenschrift 102(9):260- carcinoma of the thyroid. Clin Oncol 4:3-10.
264. 98. Bukowski RM, Brown L, Weick JK, Groppe CW, Purvis J
79. Hötling T, Meybier H, Buhr H 1990 Stellenwert der tra¬ 1983 Combination chemotherapy of metastatic thyroid can¬
cheotomie in der behandlung des respiratorischen notfalls cer: phase II study. Am J Clin Oncol 6:579-581.
beim anaplastichen schilddrüsenkarzinom. Wiener Klinische 99. Kober F, Heiss A, Keminger K, Kepisch D 1990 Chemother¬
Wochenschrift 102(9):264-266. apie hochmaligner schilddrüsentumore. Weiner Klinische
80. Levendag PC, De Porre PMZR, van Putten WLJ 1993 Wochenschrift 102(9):274-276.
Anaplastic carcinoma of the thyroid gland treated by radi¬ 100. Ekman ET, Lundell G, Tennvall J, Wallin G 1990
ation therapy. Int J Radiation Oncol Biol Phys 26(1):125- Chemotherapy and multimodality treatment in thyroid car¬
128. cinoma. Otolaryngol Clin North Am 23(3):523-527.
81. Kim JH, Leeper RD 1983 Treatment of anaplastic giant and 101. Satake S, Sugawara I, Watanabe M, Takami H 1997 Lack
spindle cell carcinoma of the thyroid gland with combina¬ of a point mutation of human DNA topoisomerase II in
tion adriamycin and radiation therapy: A new approach. multidrug-resistant anaplastic thyroid carcinoma cell lines.
Cancer 52(6):954-957. Cancer Lett 116:33-39.
82. Kim JH, Leeper RD 1987 Treatment of locally advanced 102. Loe DW, Deeley RG, Cole SPC 1996 Biology of the multi-
thyroid carcinoma with combination doxorubicin and radi¬ drug resistance-associated protein, MRP. Eur J Cancer
ation therapy. Cancer 60:2372-2375. 32A(6):945-957.
83. Wong CS, Van Dyk J, Simpson WJ 1991 Myelopathy fol¬ 103. Lehnert M 1996 Clinical multidrug resistance in cancer: a
lowing hyperfractionated accelerated radiotherapy for multifactorial problem. Eur J Cancer 32A(6):912-920.
anaplastic thyroid carcinoma. Radiother Oncol 20:3-9. 104. Venkataraman GM, Yatin M, Ain KB. MDR1 and MRP1
84. Simpson WJ 1980 Anaplastic thyroid carcinoma: a new ap¬ mRNA expression in primary thyroid cancers and cell lines.
proach. Can J Surg 23(l):25-27. 79th Annual Meeting of the Endocrine Society. Minneapo¬
85. Tennvall J, Tallroth E, El Hassan A, Lundell G, Akerman lis, MN, 1997: 248.
M, Biörklund A, Blomgren H, Löwhagen , Wallin G 1990 105. Auersperg M, Us-Krasovec M, Pétrie G, Pogacnik A, Besic
Anaplastic thyroid carcinoma: Doxorubicin, hyperfraction¬ 1990 Results of combined modality treatment in poorly
ated radiotherapy and surgery. Acta Oncol 29(8): differentiated and anaplastic thyroid carcinoma. Wien Klin
1025-1028. Wochenschr 102(9):267-270.
86. Tennvall J, Lundell G, Hallquist A, Wahlberg , Wallin G, 106. Schlumberger M, Parmentier C, Delisle M-J, Couette J-E,
726 AIN

Droz J-P, Sarrazin D 1991 Combination therapy for O, Scherbaum WA, Dralle H, Hoang-Vu C 1997 CD97: A
anaplastic giant cell thyroid carcinoma. Cancer 67(3): dedifferentiation marker in human thyroid carcinoma. Can¬
564-566. cer Res 57:1798-1806.
107. Tallroth E, Wallin G, Lundell G, Löwhagen , Einhorn J 123. Ain KB, Taylor KD, Tofiq S, Venkataraman G 1997 So-
1987 Multimodality treatment in anaplastic giant cell thy¬ matostatin receptor subtype expression in human thyroid
roid carcinoma. Cancer 60(7):1428-1431. and thyroid carcinoma cell lines. J Clin Endocrinol Metab
108. Werner B, Abele J, Alveryd A, Björklund A, Franzén S, 82(6):1857-1862.
Granberg P-O, Landberg , Lundell G, Löwhagen T, Sund- 124. Ain KB, Taylor KD 1994 Somatostatin analogs affect pro¬
blad R, Tennvall J 1984 Multimodal therapy in anaplastic liferation of human thyroid carcinoma cell lines in vitro. J
giant cell thyroid carcinoma. World J Surg 8(l):64-70. Clin Endocrinol Metab 78(5):1097-1102.
109. Serrano M, Monroy C, Rodriguez-Garcia JL, Pais JR, Fer¬ 125. Iwasa K, Noguchi M, Mori K, Ohta N, Miyazaki I, Nono-
nandez-Garrido M 1994 Tratamiento combinado del carci¬ mura A, Mizukami Y, Nakamura S, Michigishi 1995
noma anaplásico de tiroides. Ann Med Intern (Madrid) Anaplastic thyroid carcinoma producing the granulocyte
ll(6):288-290. colony stimulating factor (G-CSF): Report of a case. Surg
110. Casterline PF, Jaques DA, Blom H, Wartofsky L 1980 Today 25:158-160.
Anaplastic giant and spindle-cell carcinoma of the thyroid: 126. Yoshida M, Matsuzaki H, Sakata K, Takeya M, Kato K,
A different therapeutic approach. Cancer 45(7):1689-1692. Mizushima S, Kawakita M, Takatsuki 1992 Neutrophil
111. Sweeney PJ, Haraf DJ, Recant W, Kaplan EL, Vokes EE chemotactic factors produced by a cell line from thyroid car¬
1996 Anaplastic carcinoma of the thyroid. Ann Oncology cinoma. Cancer Res 52:464-469.
7:739-744. 127. Oka Y, Kobayashi T, Fujita S, Matsuura N, Okamoto S,
112. Yoshida A, Fukazawa M, Ushio H, Aiyoshi Y, Soeda S, Ito Asakawa H, Murata A, Mori 1993 Establishment of a
1989 Study of cell kinetics in anaplastic thyroid carci¬ human anaplastic thyroid cancer cell line secreting granu¬
noma transplanted to nude mice. J Surg Oncol 41:1—4. locyte colony-stimulating factor in response to cytokines. In
113. Gioanni J, Zanghellini E, Mazeau C, Zhang D, Courdi A, Vitro Cell Dev Biol 29A:537-542.
Farger M, Lambert JC, Duplay H, Schneider M 1991 Car- 128. Aust G, Hofmann A, Laue S, Ode-Hakim S, Scherbaum WA
actérisation d'une lignée cellulaire humaine provenant d'un 1996 Differential regulation of granulocyte-macrophage
carcinome anaplasique de la thyroïde. Bull Cancer 78:1053- colony-stimulating factor mRNA and protein expression in
1062. human thyrocytes and thyroid-derived fibroblasts by inter-
114. Boghaert ER, Ain KB, Taylor K, Greenberg VL, Fowler C, leukin-la and tumour necrosis factor- . J Endocrinol
Zimmer SG 1996 Quantitative and qualitative differences 151:277-285.
in growth, invasion, and metastasis of an anaplastic and a 129. Baba M, Kobayashi , Oka Y, Yagyu T, Tamaki Y, Takeda
papillary human thyroid cancer cell line in vitro and in vivo. T, Monden , Shimano , Tsuji , Murakami H, Mori
J Clin Exp Métastases 14(5):440-450. 1993 Preparation of a human monoclonal antibody derived
115. Ain KB, Ishizawar RC, Taylor KD 1994 Suramin inhibits from cervical lymph nodes of a patient with anaplastic car¬
growth of differentiated and anaplastic human thyroid car¬ cinoma of the thyroid. Hum Antibod Hybridomas
cinomas in monolayer and spheroid cultures with disparate 4:181-185.
effects in vivo. p. 375, 76th Annual Meeting of the En¬ 130. Asakawa H, Kobayashi T, Komoike Y, Yanagawa T, Taka¬
docrine Society. Anaheim, CA. hashi M, Wakasugi E, Maruyama H, Tamaki Y, Matsuzawa
116. Pályi I, Péter I, Daubner D, Vincze B, Lôrincz I 1993 Es¬ Y, Monden M 1996 Establishment of anaplastic thyroid car¬
tablishment, characterization and drug sensitivity of a new cinoma cell lines useful for analysis of chemosensitivity and
anaplastic thyroid carcinoma cell line (BHT-101). Virchows carcinogenesis. J Clin Endocrinol Metab 81(10):3547-3552.
Archiv Cell Pathol 63:263-269. 131. Wenisch HJC, Wagner RH, Schumm-Draeger P-M, Encke
117. Abe Y, Ichikawa Y, Muraki T, Ito K, Homma M 1981 Thy¬ A 1986 Cytostatic drug therapy in anaplastic thyroid carci¬
rotropin (TSH) receptor and adenylate cyclase activity in noma. World J Surg 10(5):762-769.
human thyroid tumors: Absence of high affinity receptor 132. Ain KB, Tofiq S, Taylor KD 1996 Antineoplastic activity of
and loss of TSH responsiveness in undifferentiated thyroid taxol against human anaplastic thyroid carcinoma cell lines
carcinoma. J Clin Endocrinol Metab 521:23-28. in vitro and in vivo. J Clin Endocrinol Metab 81(10):
118. Lazzereschi D, Ranieri A, Mincione G, Taccogna S, Nardi 3650-3653.
F, Colletta G 1997 Human malignant thyroid tumors dis¬ 133. Brown I, Carpenter RN 1991 Endogenous 211At -particle
played reduced levels of transforming growth factor ß re¬ radiotherapy for undifferentiated thyroid cancer. Acta Ra¬
ceptor type II messenger RNA and protein. Cancer Res diologica Suppl 376:174-175.
57:2071-2076. 134. Cobb LM, Harrison A, Dudley NE, Carr TEF, Humphreys
119. Heldin N-E, Gustavsson B, Claesson-Welsh L, Hammacher JA 1989 Relative concentration of astatine-211 and iodine-
A, Mark J, Heldin C- , Westermark 1988 Aberrant ex¬ 125 by human fetal thyroid and carcinoma of the thyroid
pression of receptors for platelet-derived growth factor in in nude mice. Radiother Oncol 13:203-209.
an anaplastic thyroid carcinoma cell line. Proc Nati Acad
Sci USA 85(12):9302-9306.
120. Itoh K, Fukuda K, Matsuzaki T, Takao S, Ohyama M 1989
Human endothelial cell growth factors derived from thyroid Address reprint requests to:
anaplastic cell carcinoma. Laryngoscope 99(5):533-537. Kenneth B. Ain, M.D.
121. Tanaka Y, Kubota K, Sasaki N, Takaku F, Uchimura H Division of Endocrinology and Molecular Medicine,
1991 Growth stimulating activity secreted by human Room MN 520
anaplastic thyroid carcinoma cells. Endocrinol Japan Department of Internal Medicine,
38(4):413-420. University of Kentucky
122. Aust G, Eichler W, Laue S, Lehmann I, Heldin N-E, Lotz 800 Rose Street, Lexington, KY 40536-0084

Das könnte Ihnen auch gefallen