ArticlePDF Available

Spontaneous Remission of Metastatic Lung Cancer Following Myxedema Coma--an Apoptosis-Related Phenomenon?

Authors:

Figures

Content may be subject to copyright.
Thyroid hormones and cancer: clinical studies of hypothyroidism
in oncology
Aleck H. Hercbergs
a
, Osnat Ashur-Fabian
b
and David Garfield
c
Introduction
A relationship between the thyroid gland and cancer was
first alluded to by Beatson in 1896 [1]. Much experimen-
tal, clinical and epidemiological effort has subsequently
been directed at defining and clarifying this putative and
controversial relationship. The possibility that a severe
deficiency in ambient thyroid hormone levels might have
a therapeutic impact on metastatic cancer was originally
suggested by Hercbergs and Leith [2]. In that report, a
complete, sustained regression of metastatic nonsmall
cell lung cancer (NSCLC) occurred in a man who had
lapsed into and was then successfully resuscitated from
myxedema coma. He lived 5 years without recurrence
before dying of unrelated causes. This unique event
found support in preclinical studies that revealed
that thyroid hormone deprivation slowed solid tumor
growth rates, whereas thyroid hormone supplementation
increased tumor growth rates [3 6]. If endogenous thyr-
oid hormone in euthyroid patients may in fact influence
cancer development [7], then the spontaneous or induced
hypothyroid state might be associated with a more favor-
able prognosis in cancer patients.
The possible association between thyroid hormone and
cancer may now be better understood following the
discovery of a membrane receptor for L-thyroxine
(T
4
)and3,5,3
0-triiodo-L-thyronine (T
3
)onastructural
protein of the plasma membrane, integrin avb3[8,9].
This integrin and the cell surface thyroid hormone
receptor appear to mediate the proliferative action of
the hormone on blood vessel cells and on tumor cells
[9]. Integrin avb3 is primarily expressed on rapidly
dividing cells. This recently understood molecular
mechanism of thyroid hormone action may shed light
on the numerous and controversial clinical studies of
thyroid hormone and cancer that have sporadically
appeared in the literature.
After a brief review of experimental thyroid hormone
actions relevant to cancer, we review clinical studies and
reports of outcomes in cancer patients with primary
hypothyroidism, spontaneous and iatrogenic.
Cell biology of thyroid hormones and
tumorigenesis and tumor cell proliferation
More than 20 years ago, Guernsey et al. [10] found
that removal of T
3
and T
4
from serum eliminated
X-ray-induced neoplastic transformation without modify-
ing cell survival. Moreover, addition of T
3
to thyroid
hormone-depleted medium re-established the expected
frequency of transformation. Borek et al. [11] also found
that T
3
facilitated chemical carcinogenesis. Using pro-
pylthiouracil (PTU) to induce hypothyroidism in intact
rats, Goodman et al. [12] showed that hypothyroidism
reduced the risk of breast cancer after 7,12-dimethylben-
z(a)anthracene (DMBA) exposure to 7% from 63% in
a
Department of Radiation Oncology, Cleveland Clinic,
Cleveland, Ohio, USA,
b
Translational Hematology
Oncology, Meir Medical Center, Kfar-Saba, Israel and
c
Promed Cancer Center, Shanghai, P.R. China
Correspondence to Aleck Hercbergs, Department of
Radiation Oncology, Cleveland Clinic, Cleveland,
OH 44195, USA
E-mail: hercbergs@gmail.com
Current Opinion in Endocrinology, Diabetes &
Obesity 2010, 17:432– 436
Purpose of review
To collect and assess clinical reports of a putative relationship between thyroid state
and the biology of cancers of various types.
Recent findings
A number of prospective case–control studies reviewed here have suggested that
subclinical hyperthyroidism increases risk of certain solid tumors and that spontaneous
hypothyroidism may delay onset or reduce aggressiveness of cancers. Small case
studies have reached similar conclusions. A controlled prospective trial of induced
hypothyroidism beneficially affected the course of glioblastoma. A context in which
to interpret such findings is the recent description of a plasma membrane receptor for
thyroid hormone on cancer cells and dividing tumor-associated endothelial cells.
Summary
Accumulating clinical evidence may justify new, broadly-based controlled studies in
cancer patients of the possible contribution of thyroid hormone to tumor behavior.
Keywords
cancer, hypothyroxinemia, integrin avb3, L-thyroxine, triiodothyronine
Curr Opin Endocrinol Diabetes Obes 17:432 –436
!2010 Wolters Kluwer Health | Lippincott Williams & Wilkins
1752-296X
1752-296X !2010 Wolters Kluwer Health | Lippincott Williams & Wilkins DOI:10.1097/MED.0b013e32833d9710
controls. PTU and thyroid hormone replacement resulted
in a 78% incidence of mammary cancer in DMBA-treated
animals. Such studies were not widely cited, at least in
part because the traditional concept of genomic thyroid
hormone action through nuclear receptor proteins
(thyroid hormone receptors) (see review [9]) resulted
in transcription of genes that supported homeostasis of
the activities of normal cells [13,14]. Further, there were
at the time of publication of these reports no substantial
clinical studies of thyroid hypofunction or hyperfunction
and cancer risk.
A plasma membrane receptor for thyroid hormone is the
basis for a nongenomic mechanism of hormone action
that appears to contribute to proliferation of integrin
avb3-bearing cells [9,15
!
,16]. As noted above, such cells
are usually tumor cells or dividing endothelial or vascular
smooth muscle cells. In this context, earlier animal
studies of thyroid hormone and cancer [3–6] may be
more understandable. Similarly, there are recent studies
on animals of tetraiodothyroacetic acid (tetrac), a thyroid
hormone analogue that disrupts the function of the integ-
rin avb3 receptor on human cancer xenografts and induces
tumor regression and slower growth [15
!
,16,17]. This
receptor also is apparently required for induction by thyr-
oid hormone of angiogenesis, including that relevant to
tumor support [8,9].
Cancer and thyroid function
Epidemiological studies of cancer and thyroid function.
Does hyperthyroidism increase and hypothyroidism
reduce cancer risk?
In a prospective study of almost 30 000 individuals fol-
lowed for 9 years, a low thyrotropin (TSH) level, sugges-
tive of subclinical hyperthyroidism (TSH <0.5 mU/l),
was associated with increased cancer risk specifically with
lung (hazard ratio 2.60) and prostate cancers (hazard ratio
1.96). Hypothyroid function was not associated with
cancer risk [18
!
].
In a population-based case–control study, hyperthyroid-
ism was identified as a significant ovarian cancer risk
factor, with an odds ratio (OR) of 1.8 [19].
Of 1362 breast cancer patients and 1250 controls,
women with untreated hypothyroidism or goiter had a
significantly reduced risk of breast cancer [relative risk
(RR) ¼0.3]. If they had received thyroid hormone for
fertility issues, the RR rose to 4.2. If there was a family
history of breast cancer, RR was 2.6, or if late age at first
childbirth, 2.4 [20]. Postmenopausal women with breast
cancer who had the elevated thyroid hormone and
reduced TSH levels consistent with subclinical
hyperthyroidism, as well as an increased thyroid hor-
mone/estradiol ratio, had more breast cancers than
matched controls [21].
A large population-based case control study involving
532 pancreatic cancer patients found that a history of
hyperthyroidism gave an OR of 2.1 for its development
[22].
In women with renal cell carcinoma (RCC), a statistically
significantly higher use of thyroid hormone was observed
(P¼0.041) [23].
Is the onset of cancer delayed in hypothyroid patients?
Hypothyroidism was associated with older age at cancer
(breast, lung cancers) diagnosis in several studies. In the
breast cancer group, hypothyroid patients were 7
(P<0.001) [24], 7 [25] and 6 years older (P<0.035)
[26] at diagnosis in three studies. Breast cancer incidence
was significantly lower in the hypothyroid group
(P<0.003). Tumors were also smaller in the hypothyroid
group (P<0.047) and were more likely to be localized.
Euthyroid patients were also more likely to have meta-
static disease [24].
In a case–control study of lung cancer, patients with lung
cancer and a history of thyroid hormone requirement
(¼thyroid hormone replacement) had a mean age at
diagnosis of 73 years vs. 64 years for euthyroid patients
(P¼0.0006). The thyroid hormone group median survi-
val was 14.5 vs. 11.1 months (P¼0.014) [27].
A possible explanation is that hypothyroidism takes a
number of years to evolve to a clinical requirement for
thyroid hormone supplementation, which might then
elicit progression of a pre-existing, indolent, subclinical
cancer [28]. This sequence is wholly speculative.
Does cancer chemotherapy-induced hypothyroidism
contribute to outcome?
The tyrosine kinase inhibitor, sunitinib, as used in RCC,
induces unintended hypothyroidism in up to 71% of
patients [29]. Of interest is that, in a recent study,
progression-free survival (PFS) in such patients seemed
better than that in euthyroid cohorts, even with and in
spite of T
4
supplementation (P¼0.07) [30]. However,
there are as yet no studies comparing replacement with
nonreplacement thyroid hormone so that any relationship
between sunitinib-induced hypothyroidism and tumor
behavior is speculative.
Hypothyroidism, chemoradiation therapy, disease
response and survival
Hypothyroidism may increase response rates to chemo-
therapy and radiation therapy of a variety of solid tumors
[31–36]. Certain of these observations have appeared
only in preliminary form [32,34].
Thyroid hormones and cancer Hercbergs et al. 433
434 Thyroid
Table 1 Cancer outcomes across a spectrum of thyroid functions
Thyroid function Type of research No. of cases Cancer type/disease Clinical outcome References
Spontaneous
hyperthyroid
Prospective
population study
29 691 Several malignancies Significantly higher hazard
ratios for lung and
prostate cancer vs.
significantly lower for HT
Hellevik et al.
[18
!
]
Case– control 532 Pancreas Increased risk with prior
hyperthyroidism
Ko et al. [22]
Case– control 26 pts, 22
matched
controls
Breast Subclinical hyperthyroidism
associated with more
frequent cancers
Saraiva et al.
[21]
Spontaneous
hypothyroid
Case report 1 NSCLC, metastatic ‘Spontaneous’ CR following
myxedema coma
Hercbergs and
Leith [2]
Series 28 Various solid tumors 100% response (CR and PR)
rate to radiation therapy in
chemically HT pts
Hercbergs [32]
Primary hypothyroidism-
Thyroid hormone
supplemented
Population-based 1136 pts, 1088
controls
Breast, primary Less aggressive disease in
HT group, fewer
metastases, 7 years older
age at onset, smaller
tumors
Cristofanilli
et al. [24]
Comparative
study
280 Breast, all stages 5 years older for HT Backwinkel and
Jackson [25]
Comparative
study
68, 91 matched
controls
Breast, all stages 6 years older, smaller tumors,
lower stage, lower
S phase for HT
Hercbergs
et al. [26]
Comparative
study
85, 85 matched
controls
Lung, all stages 4.3 years older, longer
survival for HT
Hercbergs
et al. [27]
Comparative
study
247, 234 matched
controls
RCC, all stages Greater use of TH in
RCC pts
Rosenberg
et al. [23]
Case report 1 Breast Apparent tumor stimulation
with TH
Hercbergs [7]
Case report/
review
1 NSCLC Apparent tumor stimulation
with TH
Hercbergs [33]
Case report 1 Anaplastic thyroid Apparent tumor stimulation
with TH, CR while clinically
HT, 10-year survival
Hercbergs
et al. [44]
Series 5 Pancreas, CRC Long-term survival while on
lower dose; TH/TH
discontinued
Hercbergs
et al. [34]
Series 176 Breast Pts taking TH before
diagnosis had
greater relapse rate,
larger tumors
Burt and Schapira
[37]
Hypothyroid -[iatrogenic]
2
0
to XRT/CHEMORX/
SURG/Biologics
Retrospective 54 RCC treated with
sunitinib
Pts becoming HT with
sunitinib and treated
with TH seemed to have
worse outcome
Sabatier 2009
et al. [30]
Retrospective 155, with 59
developing HT
HNSCC Pts developing HT seemed
to have better survival
Nelson et al. [35]
Population-based 5916 (age >65) HN (excluding thyroid,
larynx, prior HT)
Longer survival in those
developing HT
Smith et al. [36]
Phase II, subset
analysis
34 RCC, melanoma treated
with IL-2/LAK cells
Higher responses with
development of HT
Atkins et al. [39]
Phase II, subset
analysis
16 RCC, metastatic,
treated with
IL-2/LAK cells
Development of HT
correlated with
better response rate
Weijl et al. [40]
Interventional
hypothyroxinemia
Phase I– II 36 Recurrent, high-grade
gliomas made HT
with PTU
Early-onset HT associated
with improved survival
Hercbergs et al.
[41] Hercbergs
et al. [42]
Phase II 20 Recurrent, high-grade
gliomas made HT
with PTU
HT associated with
improved survival
Linetsky et al.
[43]
Recurrent disease
following [re-]
initiation of
L-thyroxine in
HT pts
Case reports 4 Breast 7/9 women given TH after
mastectomy developed
recurrence, 4 of which
were late
Burt and Schapira
[37]
Case report 1 Breast Rapid progression, death
after re-starting TH,
3þyears after being in CR
Hercbergs [7]
CR, complete response; CRC, colorectal cancer; HN, head and neck; HNSCC, head and neck squamous cell carcinoma; HT, hypothyroidism; IL-2,
interleukin 2; LAK, lymphokine-activated killer; NSCLC, nonsmall cell lung cancer; PR, partial response; pts, patients; PTU, propylthiouracil; RCC, renal
cell carcinoma; TH, thyroid hormone; XRT, radiation therapy.
Progression or relapse of disease following L-thyroxine
[T
4
] supplementation
The records of 1465 patients with breast cancer, of whom
176 had taken thyroid hormone, were reviewed. Patients
who had taken thyroid hormone for more than 2 years
within 10 years of developing breast cancer showed a
greater relapse rate compared with controls at 3 years
(43.9 vs. 18.8%, P¼0.002). Their tumors were also larger
(P¼0.01) [37].
The incidence of breast cancer was significantly higher
among patients receiving thyroid hormone in comparison
to control patients. The risk increased with duration of
use, being almost 20% in those receiving it for more than
15 years. The risk was more than three-fold higher in
nulliparous women receiving thyroid hormone, reaching
33%, whereas it was only 9.25% in those not receiving
thyroid hormone [38].
Induced hypothyroidism in cancer patients
A significantly increased rate of tumor regression (5/
7¼71%) was seen in patients with advanced RCC and
melanoma who became hypothyroid, even transiently,
from interleukin-2 (IL-2) and lymphokine-activated
killer (LAK) cell therapy, compared with euthyroid
patients (5/27 ¼19%) who did not (P<0.02) [39]. A
meta-analysis similarly found a significant correlation
of response with hypothyroidism (P¼0.001) [40].
A prospective study of PTU-induced mild hypothyroid-
ism revealed that hypothyroidism was associated with
tumor regression and a statistically significant pro-
longation of survival and time-to-progression in patients
with recurrent/progressive primary brain tumors [41].
PTU induced hypothyroidism in 36 such patients and
early onset chemical hypothyroidism (within 2 months)
was seen in 18 patients. The 6-month PFS of patients
with at least two consecutive monthly readings of serum-
free [F]T4 levels below the reference range (hypothyr-
oxinemia) was 58 vs. 0% for nonhypothyroid patients
(P<0.002). Initial FT4 decline was an independent
prognostic variable and the nadir of FT4 decline also
correlated positively with overall survival (P<0.003)
[42].
A preliminary report of recurrent glioma patients ren-
dered hypothyroid similarly found that survival was sig-
nificantly prolonged with an associated clinical improve-
ment (8/12). This led to withdrawal of steroid therapy
in two patients and dose reduction in the other six.
Responding patients also had marked decrease in seizure
activity. Median time to tumor progression (TTP) was
significantly longer in the hypothyroid group (5 vs.
2.7 months; P¼0.002) with 6-month PFS of 33 vs. 0%
in the euthyroid group [43] (see Table 1) [44].
Conclusion
Review of published clinical studies and accumulating
preliminary data in cancer patients with spontaneous,
chemically, or iatrogenically induced hypothyroidism
heightens suspicion that thyroid hormone is a permissive
factor in some patients with solid tumors. Validation or
refutation of this concept requires additional prospective,
controlled studies of induced, clinically mild hypothyr-
oidism in cancer patients. A consensus statement of the
American Thyroid Association (ATA) [45] recommends
withholding thyroid hormone replacement in asympto-
matic patients without cancer who have modest
elevation of serum TSH concentration above the refer-
ence range. This recommendation provoked controversy
[46]. Until additional information is available from pro-
spective studies of hypothyroidism and cancer, however,
we suggest it is prudent to consider the ATA consensus
recommendation on thyroid hormone replacement when
managing chemically hypothyroid patients with cancer.
References and recommended reading
Papers of particular interest, published within the annual period of review, have
been highlighted as:
!of special interest
!! of outstanding interest
Additional references related to this topic can also be found in the Current
World Literature section in this issue (p. 486).
1Beatson GT. On the treatment of inoperable cases of carcinoma of the
mamma: suggestions for a new method of treatment, with illustrative cases.
Lancet 1896; 2:104– 107.
2Hercbergs A, Leith JT. Spontaneous remission of metastatic lung cancer
following myxedema coma: an apoptosis-related phenomenon? J Natl Cancer
Inst 1993; 85:1342– 1343.
3Shoemaker JP, Dagher RK. Remissions of mammary adenocarcinoma in
hypothyroid mice given 5-fluorouracil and chloroquine phosphate. J Natl
Cancer Inst 1977; 62:1575– 1578.
4Kumar MS, Chiang T, Deodhar SD. Enhancing effect of thyroxine on tumor
growth and metastasis in syngeneic mouse tumor systems. Cancer Res
1979; 39:3515– 3518.
5Mishkin SY, Pollak R, Yalovsky MA, et al. Inhibition of local and metastatic
hepatoma growth and prolongation of survival after induction of hypothyroid-
ism. Cancer Res 1981; 41:3040– 3045.
6Theodossiou C, Skrepnik N, Robert EG, et al. Propylthiouracil-induced
hypothyroidism reduces xenograft tumor growth in athymic nude mice.
Cancer 1999; 86:1596– 1601.
7Hercbergs A. The thyroid gland as an intrinsic biologic response-modifier in
advanced neoplasia: a novel paradigm. In Vivo 1996; 10:245– 247.
8Bergh JJ, Lin H-Y, Lansing L, et al. Integrin avb3 contains a cell surface
receptor site for thyroid hormone that is linked to activation of MAPK and
induction of angiogenesis. Endocrinology 2005; 146:2864 –2871.
9Cheng SY, Leonard LJ, Davis PJ. Molecular aspects of thyroid hormone
actions. Endocr Rev 2010; 31:139–170.
10 Guernsey DL, Ong A, Borek C. Thyroid hormone modulation of X-ray-
induced in vitro neoplastic transformation. Nature (London) 1980; 288:
591– 592.
11 Borek C, Guernsey DL, Ong A, Edelman IS. Critical role played by thyroid
hormone in induction of neoplastic transformation by chemical carcinogens in
tissue culture. Proc Natl Acad Sci U S A 1983; 80:5749–5752.
12 Goodman AD, Hoekstra SJ, Marsh PS. Effects of hypothyroidism on the
induction and growth of mammary cancer induced by 7,12-dimethylben-
z(a)anthracene in the rat. Cancer Res 1980; 40:2336 –2342.
13 Feng X, Jiang Y, Meltzer P, Yen PM. Thyroid hormone regulation of hepatic
genes in vivo detected by complementary DNA microarray. Mol Endocrinol
2000; 14:947– 955.
Thyroid hormones and cancer Hercbergs et al. 435
14 Miller LD, McPhie P, Suzuki H, et al. Multitissue gene expression analysis in a
mouse model of thyroid hormone resistance. Genome Biol 5:R31.
15
!
Lin HY, Sun M, Tang HY, et al. L-Thyroxine vs. 3, 5 3-triiodo-L-thyronine
and cell proliferation: activation of mitogen-activated protein kinase and
phosphatidylinositol 3-kinase. Am J Physiol Cell Physiol 2009; 296:C980
C991.
The study reports that triiodothyronine at a physiological concentration is less
mitogenic than physiological concentrations of L-thyroxine [T
4
].
16 Yalcin M, Dyskin E, Lansing L, et al. Tetraiodothyroacetic acid (tetrac) and
nanoparticulate tetrac arrest growth of medullary carcinoma of the thyroid.
J Clin Endocrinol Metab 2010; 95:1972–1980.
17 Yalcin M, Bharali DJ, Lansing L, et al. Tetraiodothyroacetic acid (tetrac) and
tetrac nanoparticles inhibit growth of human renal cell carcinoma xenografts.
Anticancer Res 2009; 29:3825–3831.
18
!
Hellevik AI, Asvold BO, Bjøro T, et al. Thyroid function and cancer risk: a
prospective population study. Cancer Epidemiol Biomarkers Prev 2009;
18:570– 574.
First long-term prospective study of thyroid function in a large population that
identifies hyperthyroidism as a significant risk factor for the development of lung
and prostate cancers.
19 Ness RB, Grisso JA, Cottreau C, et al. Factors related to inflammation of the
ovarian epithelium and risk of ovarian cancer. Epidemiology 2000; 11:111–
117.
20 Hoffman DA, McConahey WM, Brinton LA, et al. Breast cancer in hypothyroid
women using thyroid supplements. JAMA 1984; 251:616 –619.
21 Saraiva PP, Figueirado NB, Padovani CR, et al. Profile of thyroid hormones in
breast cancer patients. Braz J Med Res 2005; 38:761–765.
22 Ko AH, Wang F, Holly EA. Pancreatic cancer and medical history in a
population-based case-control study in the San Francisco Bay Area,
California. Cancer Causes Control 2007; 18:809 –819.
23 Rosenberg AG, Dexeus F, Swanson DA, et al. Relationship of thyroid disease
to renal cell carcinoma: an epidemiologic study. Urology 1990; 35:492–
498.
24 Cristofanilli M, Yamamura Y, Kau S-W, et al. Thyroid hormone and breast
carcinoma. Primary hypothyroidism is associated with a reduced incidence of
primary breast carcinoma. Cancer 2005; 103:1122 –1128.
25 Backwinkel K, Jackson A. Some features of breast cancer and thyroid
deficiency. Cancer 1964; 17:1174–1176.
26 Hercbergs AH, Daw H, Thakur S. Primary hypothyroidism and breast cancer:
clinical and pathological risk reduction correlates from a case control study.
Proceedings of the 80th Annual Meeting of the American thyroid Association;
Palm Beach, Florida, poster no.178; 2009.
27 Hercbergs A, Mason J, Reddy C. Thyroid hormones and lung cancer: primary
hypothyroidism is prognostically significant for survival in lung cancer
[abstract #4440]. 95th Annual Meeting, AACR; Orlando, FL; 30 March 2004.
28 Huber G, Staub JJ, Meier C, et al. Prospective study of the spontaneous
course of subclinical hypothyroidism: prognostic value of thyrotrophin, thyroid
reserve, and thyroid antibodies. J Clin Endocrinol Metab 2002; 87:3221
3226.
29 Illouz F, Laboureau-Soares S, Dubois S. Tyrosine kinase inhibitors and
modifications of thyroid function. Euro J Endocrinol 2009; 160:331–
336.
30 Sabatier R, Gravis G, Deville, J. Hypothyroidism and survival during sunitinib
therapy in metastatic renal cell cancer: a prospective observational analysis
[abstract # 317]. ASCO Genitourinary Cancers Symposium; 2009.
31 Hercbergs A. Hypothyroidism and tumor regression. N Engl J Med 1988;
319:1315.
32 Hercbergs A. High tumor response rate to radiation therapy in biochemically
hypothyroid patients [abstract #167]. Proceedings of AACR; 1997. p. 248.
33 Hercbergs A. Spontaneous remission of cancer: a thyroid hormone depen-
dent phenomenon? Anticancer Res 1999; 19:4839 –4844.
34 Hercbergs A, Daw H, Ayoub W. Possible association of hypothyroidism with
longevity in advanced colorectal and pancreatic cancer: a case study. 79th
Annual Meeting of American Thyroid Association; Chicago, IL; October 2008.
Thyroid 2008; 8:S-18.
35 Nelson M, Hercbergs A, Rybicki L, et al. Association between development of
hypothyroidism and improved survival in patients with head and neck cancer.
Arch Otolaryngol Head Neck Surg 2006; 132:1041–1046.
36 Smith GL, Smith BD, Gardes AS. Hypothyroidism in older patients with head
and neck cancer after treatment with radiation: a population-based study.
Head Neck 2009; 31:1031– 1038.
37 Burt JRF, Schapira DV. Thyroid hormone: a modifier of the natural history of
breast cancer [abstract no. 738]. Annual Meeting of the American Association
for Cancer Research; 1983.
38 Kapdi CC, Wolfe JN, Shapiro S, et al. Breast cancer. Relationship to thyroid
supplements for hypothyroidism. JAMA 1976; 236:1124 –1127.
39 Atkins MB, Mier JW, Parkinson DR, et al. Hypothyroidism after treatment with
interleukin-2 and lymphokine-activated killer cells. N Engl J Med 1988;
318:1557– 1563.
40 Weijl NI, Van der Harst D, Brand A, et al. Hypothyroidism during immunother-
apy with interleukin-2 is associated with antithyroid antibodies and response
to treatment. J Clin Oncol 1993; 11:1376 –1383.
41 Hercbergs A, Goyal LK, Suh JH, et al. Propylthiouracil-induced chemical
hypothyroidism with high-dose tamoxifen prolongs survival in recurrent high
grade glioma: a phase I/II study. Anticancer Res 2003; 23 (1B):617 626.
42 Hercbergs A, Suh J, Reddy C, Goyal L. Early onset propylthiouracil-induced
hypothyroidism is associated with improved survival in recurrent high grade
glioma [abstract #1211]. 99th Annual meeting AACR; San Diego, CA; April
2008.
43 Linetsky E, Hercbergs A, Dotan S, et al. Time to tumor progression (TTP) and
quality of life (QOL) following propylthiouracil induction of chemical hypothyr-
oidism in failed malignant gliomas [abstract no. 144]. World Federation of
Neuro-Oncology Second Quadrennial Meeting; Edinburgh; 5 May 2005.
p. 40.
44 Hercbergs A, Setrakian S, Abdelmannan D. Anaplastic thyroid CA: 10-year
survival in a man with uncorrected primary hypothyroidism. Thyroid 2007; 17
(Suppl 1):S-93.
45 Surks MI, Ortiz E, Daniels GH. Subclinical thyroid disease: scientific review
and guidelines for diagnosis and management. JAMA 2004; 291:228 –238.
46 Gharib H, Tuttle RM, Baskin HJ, et al. Subclinical thyroid dysfunction: a joint
statement on management from the American Association of Clinical
Endocrinologists, the American Thyroid Association, and the Endocrine
Society. J Clin Endocrinol Metab 2005; 90:581 –585.
436 Thyroid
... The interest for the TH-cancer association was rekindled with the cloning of cDNAs encoding the nuclear TH receptors (TRs) in 1986 [5], which heralded a new realm of investigation. In 1993, the case report of spontaneous remission of metastatic NSCLC following recovery from myxedema coma by Hercbergs and Leith [6] paved the way for the increasing recognition of the implication of TH in a wide array of malignancies beyond ...
... The hallmark case report of Hercbergs and Leith in 1993, pointing to the anticancer potential of hypothyroidism in lung cancer in the clinical setting [6], launched an evolving field of research. In this case report, the authors described the remission of metastatic NSCLC, previously treated with chemotherapy and radiotherapy, after recovery of myxedema coma induced by amiodarone in a 69-year-old male patient [6]. ...
... The hallmark case report of Hercbergs and Leith in 1993, pointing to the anticancer potential of hypothyroidism in lung cancer in the clinical setting [6], launched an evolving field of research. In this case report, the authors described the remission of metastatic NSCLC, previously treated with chemotherapy and radiotherapy, after recovery of myxedema coma induced by amiodarone in a 69-year-old male patient [6]. The myxedema coma was initially treated with high doses of levothyroxine (LT4). ...
Article
Full-text available
Exemplifying the long-pursued thyroid hormones (TH)–cancer association, the TH–lung cancer association is a compelling, yet elusive, issue. The present narrative review provides background knowledge on the molecular aspects of TH actions, with focus on the contribution of TH to hallmarks of cancer. Then, it provides a comprehensive overview of data pertinent to the TH–lung cancer association garnered over the last three decades and identifies obstacles that need to be overcome to enable harnessing this association in the clinical setting. TH contribute to all hallmarks of cancer through integration of diverse actions, currently classified according to molecular background. Despite the increasingly recognized implication of TH in lung cancer, three pending queries need to be resolved to empower a tailored approach: (1) How to stratify patients with TH-sensitive lung tumors? (2) How is determined whether TH promote or inhibit lung cancer progression? (3) How to mimic the antitumor and/or abrogate the tumor-promoting TH actions in lung cancer? To address these queries, research should prioritize the elucidation of the crosstalk between TH signaling and oncogenic signaling implicated in lung cancer initiation and progression, and the development of efficient, safe, and feasible strategies leveraging this crosstalk in therapeutics.
... In the following decades a variety of studies focused on what has been called hormonal carcinogenesis [235]. In particular, between the end of the 1970s and the beginning of the 1990s, different studies, performed both in culture and in vivo, suggested that THs had a role in neoplastic transformation, and that hypothyroidism could reverse TH-dependent growth and spread of tumor cells [236][237][238][239][240][241]. For example, a patient affected by a metastatic lung cancer, during chemotherapy, began suffering of cardiac arrhythmia and because of this was also treated with amiodarone HCl. ...
... The patient was treated with thyroxine and recovered. Surprisingly, the tumor mass was reduced and the patient lived for four years after the coma event [239]. Similarly, the experimental induction of a hypothyroid state in mice affected by either spontaneous or xenografted human tumors was often shown to reduce the rate of cancer growth [239]. ...
... Surprisingly, the tumor mass was reduced and the patient lived for four years after the coma event [239]. Similarly, the experimental induction of a hypothyroid state in mice affected by either spontaneous or xenografted human tumors was often shown to reduce the rate of cancer growth [239]. Moreover, it has been reported that high free T4 (FT4) levels are associated with an increased risk of any solid, lung, and breast cancer [242,243], and that hypothyroidism is associated with an older age of onset of different kinds of cancer (for example, breast and lung cancers) [244][245][246]. ...
Article
Full-text available
Simple Summary Development and function of the mammalian brain clearly require precise regulation of gene expression at both the transcriptional and post-transcriptional level. Thyroid hormones have been recognized to play a fundamental role in these processes, by acting at multiple levels and in different brain cell types, through direct effects on transcription, mediated by nuclear receptors, and also by triggering transduction pathways at the plasma membrane. At the same time, due to their effects on proliferation, differentiation, and cell metabolism, thyroid hormones may have a critical role in different kinds of cancer, including brain cancer. Abstract The development and maturation of the mammalian brain are regulated by thyroid hormones (THs). Both hypothyroidism and hyperthyroidism cause serious anomalies in the organization and function of the nervous system. Most importantly, brain development is sensitive to TH supply well before the onset of the fetal thyroid function, and thus depends on the trans-placental transfer of maternal THs during pregnancy. Although the mechanism of action of THs mainly involves direct regulation of gene expression (genomic effects), mediated by nuclear receptors (THRs), it is now clear that THs can elicit cell responses also by binding to plasma membrane sites (non-genomic effects). Genomic and non-genomic effects of THs cooperate in modeling chromatin organization and function, thus controlling proliferation, maturation, and metabolism of the nervous system. However, the complex interplay of THs with their targets has also been suggested to impact cancer proliferation as well as metastatic processes. Herein, after discussing the general mechanisms of action of THs and their physiological effects on the nervous system, we will summarize a collection of data showing that thyroid hormone levels might influence cancer proliferation and invasion.
... However, the impact of hypothyroidism on the risk and/or the prognosis of solid non-thyroid cancer is not a consistent finding. Such inconclusive data hamper, so far, the exploitation of hypothyroidism as an anticancer strategy despite initial enthusiasm about the anticancer efficacy of myxedema coma in metastatic non-small-cell lung cancer (NSCLC), arising from a hallmark case report in 1993 [35]. ...
... The hallmark case report on the remission of metastatic NSCLC ascribed to amiodaroneinduced myxedema coma by Hercbergs and Leith in 1993 [35] launched an evolving field of clinical research on the anticancer potential of hypothyroidism. In 2010, a review of the published up-to-that-time clinical studies indicated that hypothyroidism of any cause-i.e., spontaneous or iatrogenic-is an omen of favorable cancer prognosis thanks to inhibition of the "permissive role of THs" in cancer [93]. ...
Article
Full-text available
Hypothyroidism in patients with solid non-thyroid cancer is a tantalizing entity, integrating an intriguing thyroid hormones (THs)–cancer association with the complexity of hypothyroidism itself. The present narrative review provides a comprehensive overview of the clinical relevance of hypothyroidism in solid non-thyroid cancer. Hypothyroidism in patients with solid non-thyroid cancer is reminiscent of hypothyroidism in the general population, yet also poses distinct challenges due to the dual role of THs in cancer: promoting versus inhibitory. Close collaboration between oncologists and endocrinologists will enable the prompt and personalized diagnosis and treatment of hypothyroidism in patients with solid non-thyroid cancer. Clinical data indicate that hypothyroidism is a predictor of a decreased or increased risk of solid non-thyroid cancer and is a prognostic factor of favorable or unfavorable prognosis in solid non-thyroid cancer. However, the impact of hypothyroidism with respect to the risk and/or prognosis of solid non-thyroid cancer is not a consistent finding. To harness hypothyroidism, or THs replacement, as a personalized anticancer strategy for solid non-thyroid cancer, four prerequisites need to be fulfilled, namely: (i) deciphering the dual THs actions in cancer; (ii) identifying interventions in THs status and developing agents that block tumor-promoting THs actions and/or mimic anticancer THs actions; (iii) appropriate patient selection; and (iv) counteracting current methodological limitations.
... T3-directed gene expression in cells requires primary interactions between T3 and its nuclear receptor proteins (TRs) (1,2). T3 also regulates mitochondrial respiration (3). Because its biological half-life is significantly longer than that of T3 (6), T4 is the most commonly prescribed form of thyroid hormone replacement for clinical hypothyroidism and for suppression of endogenous pituitary thyrotropin (TSH) (1). ...
... A rare spontaneous regression of metastatic lung cancer has occurred whereby T4 and T3 became depleted to life threatening levels with myxedema coma (3). ...
Article
Full-text available
Hypothyroidism has been reported to improve survival in cancer patients but only recently has the putative mechanism been identified as a receptor for thyroxine and tri-iodothyronine on integrin αvβ3. Recognition of divergence of action of the pro-oncogenic L-thyroxine (T4) from pro-metabolic 3,5,3′-triiodo-L-thyronine (T3) has enabled clinical implementation whereby exogenous T3 may replace exogenous (or endogenous) T4 to maintain clinical euthyroid hypothyroxinemia that results in significantly better survival in advanced cancer patients without the morbidity of clinical hypothyroidism.
... As the thyroid hormones are the key regulators of several cellular processes of cell proliferation, differentiation, apoptosis, and metabolism, their association with the cancer mass has been suggested very earlier [52]. The clinical observations have supported the notion that hormonal deficiency and hypothyroidism inhibit tumor growth [24,53,54]. These actions are facilitated through several nongenomic pathways, which include activation of integrin ανβ3. ...
Article
Full-text available
The integrins belong to the cell-surface polypeptide family and are the mediating partners among the cells, and extracellular matrix (ECM). They are also involved in the biological processes of cell migration, wound healing, blood clotting, immunological response generation, tissue morphogenesis, leucocyte reticulations, and angiogenesis and are therefore very relevant in stem cell technology and are useful as biomarkers, diagnostic probes, and drug-target ligands. The ανβ3 ( alpha-nu-beta 3) integrin antagonists are an excellent target example for designing and developing newer drug candidates, drug leads and templates for various diseases, and physiological malfunctioning, including cancers. The current review examines the ανβ3 integrin structural features involved in the drug design and its antagonistic ligands and highlights the development of anti-ανβ3 integrin-antagonists as nano-architectural design-based nanomedicine, especially for cancer chemotherapy. The perspectival review discusses the ανβ3 integrin structure, mode of action, involved pathways, and the concepts utilized in nanomedicine design, and ligands related to integrins. It also covers the latest thyrointegrin approaches toward the development of anti-angiogenesis agents and entails the anti-angiogenesis approach to cancer growth inhibition through targeting by the anti-integrin ligands and related chemical entities. The current perspective on the nano-architectural design approach for the known anti-integrin compounds is also outlined.
... • Down-regulation of tumour proliferation pathways [59] Intrinsic regression • Upregulation of apoptotic pathways [60] • Return of tumour cell differentiation [61] • Removal of carcinogen [62] Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH ("Springer Nature"). Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users ("Users"), for smallscale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. ...
Article
Full-text available
Background: ALK-rearrangement is observed in < 5% non-small cell lung cancer (NSCLC) cases and prior to the advent of oral tyrosine kinase inhibitors, the natural history of oncogenic NSCLC was typically poor. Literature relating to regression of treatment-naïve NSCLC is limited, and regression without treatment has not been noted in the ALK-rearranged sub-population. Case presentation: A 76 year old 'never smoker' female with an ALK-rearranged left upper lobe T2 N0 NSCLC experienced a stroke following elective DC cardioversion for new atrial fibrillation. Following a good recovery, updated imaging demonstrated complete regression of the left upper lobe lesion and a reduction of the previously documented mediastinal lymph node. Remaining atelectasis was non-avid on repeat PET-CT imaging, 8 months from the baseline PET-CT. When the patient developed new symptoms 6 months later a further PET-CT demonstrated FDG-avid local recurrence. She completed 55 Gy in 20 fractions but at 18 months post-radiotherapy there was radiological progression in the lungs with new pulmonary metastases and effusion and new bone metastases. Owing to poor performance status, she was not considered fit for targeted therapy and died 5 months later. Conclusion: All reported cases of spontaneous regression in lung cancer have been collated within. Documented precipitants of spontaneous regression across tumour types include biopsy and immune reconstitution; stroke has not been reported previously. The favourable response achieved with radical radiotherapy alone in this unusual case of indolent oncogenic NSCLC reinforces the applicability of radiotherapy in locally advanced ALK-rearranged tumours, in cases not behaving aggressively. As a common embolic event affecting the neurological and pulmonary vasculature is less likely, an immune-mediated mechanism may underpin the phenomenon described in this patient, implying that hitherto unharnessed principles of immuno-oncology may have relevance in oncogenic NSCLC. Alternatively, high electrical voltage applied percutaneously adjacent to the tumour during cardioversion in this patient may have induced local tumour cell lethality.
... However, other epidemiological studies e.g. in patients with ovarian cancer reported conflicting results suggesting that hyperthyroidism and overall history of hypothyroidism are associated with worse 5-year survival [11]. A case report of a patient suffering from metastatic nonesmall-cell lung cancer surviving a myxoedema coma and subsequent stabilization of cancer progression for more than 5 years, was the basis for clinical attempts to interfere with TH signalling in patients with cancer [12]. Chemical hypothyroidism was induced successfully using the antithyroid thioamide, i.e. propylthiouracil, followed by carboplatin chemotherapy or tamoxifen therapy in patients with glioblastoma, however efficacy data are not yet sufficiently available to evaluate clinical activity [13,14]. ...
Article
Full-text available
Background Several preclinical and epidemiologic studies have indicated tumour-promoting effects of thyroid hormones (THs). However, very limited knowledge exists on the prognostic impact of thyroid function in metastatic cancer. Methods We compiled a discovery cohort of 1692 patients with newly diagnosed brain metastases (BMs) of solid cancers treated at the Medical University of Vienna and an independent validation cohort of 191 patients with newly diagnosed BMs treated at the University Hospital Zurich. Results Hypothyroidism before diagnosis of cancer was evident in 133 of 1692 (7.9%) patients of the discovery, and in 18 of 191 (9.4%) patients of the validation cohort. In the discovery cohort, hypothyroidism was statistically significantly associated with favourable survival prognosis from diagnosis of cancer (31 vs. 21 months; p = 0.0026) and with survival prognosis from diagnosis of BMs (12 vs. 7 months; p = 0.0079). In multivariate analysis including the diagnosis-specific graded prognostic assessment score, primary tumour type and sex, hypothyroidism was an independent factor associated with survival after diagnosis of BMs (hazard ratio: 0.76; 95% confidence interval [CI]: (0.63; 0.91; p = 0.0034). In the validation cohort, the association of hypothyroidism and favourable survival prognosis from diagnosis of cancer (55 vs. 11 months; p = 0.00058), as well as from diagnosis of BMs (40 vs. 10 months; p = 0.0036) was confirmed. Conclusion Pre-existing hypothyroidism was strongly and independently associated with prognosis in patients with newly diagnosed BMs, supporting the evidence from preclinical data that THs may indeed have a tumour-promoting effect. Further investigation of the underlying pathobiological mechanism and potential therapeutic implications are required.
... Considering the reported antitumor impact of the hypothyroid milieu [161], the correction of ir hypothyroidism is challenging. The presumed tumor promoting role of thyroid hormones has been attributed to local intracellular elevations of thyroid hormones, which activate pro-proliferative integrin αvβ3/PI3K/ERK1/2 pathways (nongenomic actions), and to attenuation of the antitumor immunity by the extracellular hypothyroid status [162]. ...
Article
Designated as scientific breakthrough of current decade, immune checkpoint inhibitors attenuate the cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and the programmed cell death 1 (PD-1)/ligand 1 (PD-L1) pathways, depriving cancer cells of a key strategy of evasion from immunosurveillance. The reinvigoration of immune response translates into clinical success, inevitably entwined with a novel constellation of immune-related adverse events. The present review dissects the endocrine immune-related adverse events, emphasizing their unique profile featured by unpredictable onset, irreversibility, nonspecific symptoms, wide clinical spectrum and sophisticated diagnostic work-up. Guidelines advocate individualized decision-making process guided by clinicians' judgement. Future perspective should be governed by five principles – prevention, anticipation, detection, treatment, monitoring – aiming to gain the optimal profit diminishing immunotoxicity.
... The levels of thyroid hormone are likely to be involved in regulating proliferation, invasion, and metastasis of tumors, ultimately affecting the prognosis of patients with cancer. 5 Thyroid hormones can promote cell division and inhibit cell apoptosis, 2 and the physiological concentration of thyroid hormones can promote angiogenesis. 6 Thyroid hormones might be mediated by phosphatidylinositol-3-kinase and the mitogen activated protein kinase pathway, and involve stimulation of angiogenesis via the avb3 pathway. ...
Article
Full-text available
Lung adenocarcinoma is a form of non-small-cell lung cancer with high mortality in the advanced stages, and is one of the most common histological subtypes of lung cancer in most countries. Prognosis of lung adenocarcinoma is generally poor, with a median survival of 4–13 months. We report a case of unusually prolonged survival of a patient with advanced lung adenocarcinoma complicated by hypothyroidism. A 71-year-old man with stage IV lung adenocarcinoma presented with hypothyroidism. Surprisingly, without any anti-tumor and anti-hypothyroidism therapy, he survived this lung cancer for longer than 2.5 years before his last follow-up visit. Patients with advanced lung adenocarcinoma rarely survive for longer than 2 years, even after therapy. We hypothesize that hypothyroidism is the cause for this discrepancy. Thyroid hormones can promote growth of carcinoma. Therefore, hypothyroidism appears to be beneficial to anti-cancer therapy. We believe that hypothyroidism, as an adverse event commonly occurring in anti-tumor therapy (e.g., an immune checkpoint inhibitor), might not be able to be completely eliminated.
Article
Hypothyroidism in the setting of cancer is a puzzling entity due to the dual role of the thyroid hormones (TH) in cancer – promoting versus inhibitory – and the complexity of the hypothyroidism itself. The present review provides a comprehensive overview of the personalized approach to hypothyroidism in patients with solid nonthyroid cancer, focusing on current challenges, unmet needs and future perspectives. Major electronic databases were searched from January 2011 until March 2022. The milestones of the refinement of such a personalized approach are prompt diagnosis, proper TH replacement and development of interventions and/or pharmaceutical agents to exploit hypothyroidism or, on the contrary, TH replacement as an anticancer strategy. Further elucidation of the dual role of TH in cancer – especially of the interference of TH signaling with the hallmarks of cancer – is anticipated to inform decision-making and optimize patient selection.
Article
Full-text available
Spontaneous remission (SR) of neoplasia is a rare biological event. Very few reports provide evidence for an eliciting event or mechanism. The only case in the literature of SR of lung cancer following myxedema coma is suggested to have been an instance of thyroid hormone deprivation-induced total tumor apoptosis. Review of the collective data suggests that the thyroid hormones modulate pleiotropic neoplasia--abetting mechanisms and that hypothyroidism may enhance the predisposition of neoplasms to spontaneous and therapy induced regression by lowering thresholds for apoptosis.
Article
The direct oncogenic potential of X rays has been demonstrated by the in vitro neoplastic transformation of mammalian cells in culture, a technique which permits the study of oncogenesis in the absence of host-specific effects. Although several agents are known to modulate in vitro neoplastic transformation by X rays, little is known of the effects of hormones. We now describe experiments which show that the presence of thyroid hormone is necessary for in vitro neoplastic transformation by X rays in cells of an established mouse fibroblast culture (C3H/10T1/2) and in early-passage diploid hamster embryo cells.
Article
In two separate experiments, treatment of C3H/He mice bearing transplantable mammary adenocarcinomas (C3HBA) with a regimen of 6-propylthiouracil (PTUra) and 5-fluorouracil (FUra) plus chloroquine phosphate (CP) resulted in complete remissions of 77 and 65%, respectively. Treatment with PTUra alone resulted in 41% remissions in experiment 1 and 35% remissions in experiment 2. None of the nontreated control mice in either experiment had spontaneous remissions, and all controls died in each experiment. The principal effect was apparently due to the treatment with PTUra, inasmuch as most of the tumors disappeared during the 21-day treatment period. This observation indicated that the proper timing of the thyroid treatment with PTUra was crucial to achieve the best results. The combined FUra+CP regimen augmented the effects of the thyroid treatment and resulted in an increase in remissions.
Article
This study was undertaken to determine the relationship between thyroid supplements and breast cancer. The incidence of breast cancer among the patients who received thyroid supplements was 12.13%, while in the control group it was 6.2%. The incidence rate of breast cancer was 10%, 9.42%, and 19.48% among patients who received thyroid supplements for one to five, 5 to 15, and for more than 15 years, respectively. The incidence of breast cancer among nulliparous women who received thyroid supplements was 33%, while in the nulliparous women without thyroid supplements the incidence was only 9.25%. Even in a specific age group, the incidence rate of breast cancer was higher among patients receiving thyroid supplements, when compared to the control patients in the same age group.
Article
Thyroid hormone participates in numerous cellular functions besides thermogenesis and metabolism. Several studies, including the recent identification of the product of an oncogene, c-erb-A, as a thyroid-hormone receptor, have shown possible involvement of thyroid hormone in the process of carcinogenesis. A recent anecdotal observation of an unusually high incidence of thyroid dysfunction in women with renal cell carcinoma led to a retrospective review of the incidence and distribution of thyroid disorders in women with renal cell carcinoma compared with a control group of women with transitional cell carcinoma of the renal pelvis, ureter, bladder, or urethra. Women with renal cell carcinoma had a statistically significantly higher percentage of hypothyroidism, thyroid disease in general, and the use of thyroid-hormone supplements as compared with the control group (P = 0.033, P = 0.005, P = 0.041, respectively). The nature of the relationship, however, could not be determined. These findings add a new dimension to renal cell carcinoma, and prospective studies are encouraged to define the contribution of thyroid hormone to renal cell carcinogenesis.
Article
The development of a goiter and hypothyroidism in a 28-year-old man in whom metastatic melanoma had been treated with interleukin-2 and lymphokine-activated killer cells (LAK cells) prompted us to assess thyroid function in patients undergoing this therapy. Thirty-four patients with advanced neoplasms who had received interleukin-2 and LAK cells were followed for at least four weeks after treatment. Seven patients (21 percent) had laboratory evidence of hypothyroidism, with a decline in the serum thyroxine concentration to below normal (less than or equal to 35 nmol per liter; normal, 65 to 148), a decline in the serum free thyroxine index, and a rise in the serum thyrotropin concentration (peak values, 7.2 to 166 mU per liter; normal, 0.5 to 5.5) 6 to 11 weeks after treatment. Two patients had elevated serum thyrotropin levels before treatment, which increased further after treatment. In two patients, these abnormal values returned to normal within 10 months. All five symptomatic patients had borderline or elevated serum antimicrosomal antibody titers after treatment; two had serum antibodies to thyroglobulin. Five of the seven patients with hypothyroidism (71 percent) but only 5 of the 27 euthyroid patients (19 percent) had evidence of tumor regression (P less than 0.02). None of 11 patients treated with interleukin-2 but not LAK cells had hypothyroidism. We conclude that treatment with interleukin-2 and LAK cells can cause hypothyroidism, possibly by exacerbating preexisting autoimmune thyroiditis, and that it may be associated with a favorable tumor response.
Article
Incubation of primary cultures of hamster embryo cells (HEC) or mouse fibroblasts (C3H/10T1/2 cells) in media depleted of thyroid hormones does not alter cell growth or survival but renders the cells resistant to neoplastic transformation by benzo[a]pyrene (B[a]P) or N-methyl-N'-nitro-N-nitrosoguanidine (MNNG), carcinogens which yield transformation rates of 10(-4)-10(-2) in media supplemented with triiodothyronine (T3). In C3H/10T1/2 cells, the times of addition or removal of the hormone indicate that T3 exerts maximum effect when added 12 hr prior to treatment with B[a]P and that the progression of transformation from the time of initiation by the carcinogen to full expression and the appearance of transformed foci was independent of the presence or absence of the hormone in the medium. Dependence of transformation on T3 concentration in the medium was observed over the physiological range of 1 pM to 100 nM in C3H/10T1/2 cells treated with B[a]P. These results were similar to our previous findings on the T3 dose-related induction of radiogenic transformation and of Na+,K+-ATPase activity. The latter effect was used as a measure of T3 induction of protein synthesis. A further indication of the potential involvement of protein synthesis in T3 action is the suppression of T3- and B[a]P-dependent transformation by cycloheximide at concentrations that inhibit protein synthesis by approximately equal to 50% in the C3H/10T1/2 cells. We suggest that thyroid hormone induces the synthesis of a host protein that plays a key role in neoplastic transformation by direct-acting chemical carcinogens and by those requiring metabolic activation. In our previous studies, similar T3-dependent mechanisms were implicated in radiogenic transformations.
Article
The risk of breast cancer associated with thyroid supplementation for iatrogenic hypothyroidism was investigated in women treated for hyperthyroidism at the Mayo Clinic, Rochester, Minn. One or more years after the diagnosis of hypothyroidism, there were 47 cases of breast cancer observed among 1,665 women compared with 52.9 cases expected, a ratio of observed to expected cases (standardized incidence ratio [SIR]) of 0.9. There was also no breast cancer excess among women who remained euthyroid (SIR = 1.0). Breast cancer risks were not influenced by various factors associated with the management of hyperthyroidism or subsequent hypothyroidism. No increased risk was observed in hypothyroid women categorized by breast cancer risk factors except for women who reported a previous breast biopsy, an excess also observed among euthyroid patients. The results of this study indicate that use of thyroid supplements does not increase the risk of breast cancer developing.
Article
We have studied the effects of hypothyroidism on the induc tion phase and growth phase of mammary cancer induced in the rat by 7,12-dimethylbenz(a)anthracene (DMBA). Moderate hypothyroidism was initiated before administration of DMBA by treatment with propylthiouracil (PTU) (0.5 to 2.0 mg/100 ml), and PTU was continued for 4 days after DMBA. Four months later, the tumor incidence in this group, 89 of 116 (77%), did not differ significantly from that in the control group, 81 of 115 (70%). Inasmuch as it is very probable that tumor induction occurs within 3 days after DMBA administration, it appears that hypothyroidism does not affect induction. When moderate hy pothyroidism was initiated by administering PTU starting 4 days after DMBA, the final tumor incidence was 64 of 121 (53%) as compared to 82 of 119 (69%) in the control rats (p < 0.01 ), but in two similar, smaller studies we did not find an effect of hypothyroidism on tumor incidence. Thus, it appears that hy pothyroidism does not enhance, and may suppress, promotion or growth of DMBA-induced cancer. When hypothyroidism was initiated before the DMBA dose and maintained continually thereafter, the final tumor incidence was 70 of 118 (59%), which did not differ significantly from the incidence in the controls of 82 of 119 (69%). Severe hypothyroidism was pro duced by administering PTU (4.0 mg/100 ml) before DMBA and continuing it thereafter, and the final tumor incidence was 3 of 45 (7%) as compared to 68 of 108 (63%) in the controls (p < 0.01). However, rats given the same PTU regimen, but maintained euthyroid by administration of thyroid hormone, had a tumor incidence of 35 of 45 (78%), indicating that PTU per se does not inhibit tumorigenesis. Moderate hypothyroid ism produced by iodine deficiency or by PTU did not affect the tumor latent period, and moderate PTU-induced hypothyroid ism did not affect the histological differentiation or estrogen dependence of the tumors. Overall, this study provides strong evidence against previous claims that hypothyroidism en hances DMBA-induced carcinogenesis.