ArticlePDF Available

Insulin-induced enhancement of antitumoral response to methotrexate in breast cancer patients

Authors:
  • Interdoctors, Uruguay

Abstract and Figures

It has been reported that insulin increases the cytotoxic effect in vitro of methotrexate by as much as 10,000-fold. The purpose of this study was to explore the clinical value of insulin as a potentiator of methotrexate. Included in this prospective, randomized clinical trial were 30 women with metastatic breast cancer resistant to fluorouracil + Adriamycin + cyclophosphamide and also resistant to hormone therapy with measurable lesions. Three groups each of ten patients received two 21-day courses of the following treatments: insulin + methotrexate, methotrexate, and insulin, respectively. In each patient, the size of the target tumor was measured before and after treatment according to the Response Evaluation Criteria In Solid Tumors. The changes in the size of the target tumor in the three groups were compared statistically. Under the trial conditions, the methotrexate-treated group and the insulin-treated group responded most frequently with progressive disease. The group treated with insulin + methotrexate responded most frequently with stable disease. The median increase in tumor size was significantly lower with insulin + methotrexate than with each drug used separately. Our results confirmed in vivo the results of previous in vitro studies showing clinical evidence that insulin potentiates methotrexate under conditions where insulin alone does not promote an increase in tumor growth. Therefore, the chemotherapy antitumoral activity must have been enhanced by the biochemical events elicited in tumor cells by insulin. In multidrug-resistant metastatic breast cancer, methotrexate + insulin produced a significant antitumoral response that was not seen with either methotrexate or insulin used separately.
Content may be subject to copyright.
ORIGINAL ARTICLE
Eduardo Lasalvia-Prisco Æ Silvia Cucchi
Jesu
´
sVa
´
zquez Æ Eduardo Lasalvia-Galante
Wilson Golomar Æ William Gordon
Insulin-induced enhancement of antitumoral response
to methotrexate in breast cancer patients
Received: 31 March 2003 / Accepted: 29 August 2003 / Published online: 4 December 2003
Springer-Verlag 2003
Abstract Purpose: It has been reported that insulin in-
creases the cytotoxic effect in vitro of methotrexate by as
much as 10,000-fold. The purpose of this study was to
explore the clinical value of insulin as a potentiator of
methotrexate. Patients and methods: Included in this
prospective, randomized clinical trial were 30 women
with metastatic breast cancer resistant to fluorouracil +
Adriamycin + cyclophosphamide and also resistant to
hormone therapy with measurable lesions. Three groups
each of ten patients received two 21-day courses of the
following treatments: insulin + methotrexate, metho-
trexate, and insulin, respec tively. In each patient, the size
of the target tumor was measured before and after
treatment according to the Response Evaluation Criteria
In Solid Tumors. The changes in the size of the target
tumor in the three groups were compared statistically.
Results: Under the trial conditions, the methotrexate-
treated group and the insulin-treated group responded
most frequently with progressive disease. The group
treated with insulin + methotrexate responded most
frequently with stable disease. The median increase in
tumor size was significantly lower with insulin +
methotrexate than with each drug used separately.
Discussion: Our results confirmed in vivo the results of
previous in vitro studies showing clinical evidence that
insulin potentiates methotrexate under conditions where
insulin alone does not promote an increase in tumor
growth. Therefore, the chemotherapy antitumoral
activity must have been enhanced by the biochemical
events elicited in tumor cells by ins ulin. Conclusions:In
multidrug-resistant metastatic breast cancer, metho-
trexate + insulin produced a significant antitumoral
response that was not seen with either methotrexate or
insulin used separately.
Keywords Breast Cancer Æ Chemotherapy Æ Insulin Æ
Methotrexate Æ Tumor growth
Introduction
It is known that slowly growing cancers have tumor cell
populations with a low-growth fraction and are less
sensitive to chemotherapy than rapidly growing tumors
with high-growth fractions [11]. Slowly growing malig-
nancies have relatively more cells in a noncycling status
and fewer cells in a cycling status than rapidly growing
malignancies. It has been demonstrated that insulin as a
pharmacological agent induces the switch from a non-
cycling to a cycling status in tumor cells [5]. In MCF-7
human breast cancer cells, insulin has been shown to
increase the cytotoxic effect of methotrexate up to
10,000-fold in vitro [1]. Ellipticine uptake is also
increased by ins ulin [9]. It has been suggested that
insulin is effective in potentiating most chemotherapy
drugs. This insulin-induced potentiation has been
Cancer Chemother Pharmacol (2004) 53: 220–224
DOI 10.1007/s00280-003-0716-7
E. Lasalvia-Prisco
Department of Medicine, School of Medicine,
University of Uruguay, Montevideo, Uruguay
S. Cucchi Æ J. Va
´
zquez Æ E. Lasalvia-Galante Æ W. Golomar
E. Lasalvia-Prisco
Interdoctors Medical Center,
Montevideo, Uruguay
E. Lasalvia-Prisco Æ J. Va
´
zquez Æ E. Lasalvia-Galante
W. Golomar
National Cancer Institute,
Montevideo, Uruguay
S. Cucchi Æ W. Gordon
PharmaBlood Inc,
North Miami Beach, Florida, USA
E. Lasalvia-Prisco (&)
(Former Director of the Department of Medicine,
School of Medicine, University of Uruguay,
Montevideo, Uruguay and National Cancer Institute,
Montevideo, Uruguay),
Research & Development Department,
PharmaBlood Inc, 2050 NE 163rd Street,
2nd Fl, 202, North Miami Beach,
Florida, 33162, USA
E-mail: research@pharmablood.com
Tel.: +1-305-9442544
Fax: +1-305-9445244
proposed as a strategy for breast cancer treatment, but
confirmatory clinical trials are still lacking [2]. This
study was carried out to confirm insulin-induced clinical
potentiation of the antitumoral effect of methotrexate as
suggested by preclinical studies and to establish a
mechanism of action for this antitumoral effect.
Patients and methods
Patients
The study was conducted in 30 patients with breast cancer admitted
to medical centers that reported medical data to the Cooperative
Trials Center (CTC) of PharmaBlood, R&D Department, Florida.
A prospective, randomized trial was carried out. All patients met
the following eligibility criteria: histologically confirmed breast
carcinoma, metastatic stage (M1); Eastern Cooperative Oncology
Group (ECOG) performance status (PS) £ 2; age £ 74 years; and
adequate hematological function (WBC count 4000/ll, neutrophil
count 2000/ll, hemoglobin level 9.0 g/dl, platelet count
10·10
4
/ll), renal function (serum creatinine £ 1.5 mg/dl, 24-h
creatinine clearance £ 60 ml/min), liver function (total bilirubin
£ 2.0 mg/dl, serum transaminases not more than twice the upper
limit of the normal range), and respiratory function (PaO
2
60 Torr). The patients included had measurable lesions, as
required by the Response Evaluation Criteria In Solid Tumors
(RECIST) system of tumor assessment [13], and if they had a
positive estrogen receptor status, they had been treated with and
become resistant to hormone therapy.
All patients included in the study had progressive disease
(RECIST criteria) after chemotherapy with at least four series of
fluorouracil + Adriamycin + cyclophosphamide (FAC) and had
not been treated with any other chemotherapy. They were ran-
domly allocated to three groups of ten patients each: group 1 was
treated with insulin + methotrexate as described below, group 2
was treated with methotrexate without insulin, and group 3 was
treated with insulin without methotrexate. Written informed con-
sent, including detailed information about risks and benefits, was
approved and signed by all the patients included in the study.
Central computerized remote randomization was performed, with
patients being allocated to one of the groups through random se-
quence generation by the permuted block method. An assessment
of the results after 30 patients had completed the trial showed that
this sample size was enough. The patients were recruited from two
oncological medical centers in Montevideo, Uruguay (first at the
National Cancer Institute and then at Interdoctors Medical Cen-
ter), both of which participated with their data in the network
operated and sponsored by the Cooperative Trials Center (CTC) of
PharmaBlood R&D Department.
The institutional ethics committee of PharmaBlood and the
institutional review boards of the participating medical centers
approved the trial. The ethical reviewers considered that an 8-week
delay before starting second-line chemotherapy after FAC had
failed in all the patients included in the trial was acceptable. This
determination was consistent with the standard of care in this
clinical situation which has been recently well summarized [3]:
Despite almost 30 years of clinical cancer research, the true
impact of second and subsequent lines of chemotherapy on the
outcome of metastatic breast cancer patients, especially on the
duration of survival, is still unknown. In the virtually incurable
metastatic setting, issues like quality of life and patients pref-
erences gain particular relevance.
The accepted protocol was resubmitted to the committee for
review in order to obtain approval for treatment of patients with
insulin alone considering the potentially harmful effect through the
activation of receptors for insulin/insulin-like growth factors. The
committee confirmed the approval on the basis of reports of no
harmful effect of this treatment [6, 7]. The results of the study
confirmed the committees criteria because no significant differences
were found in tumor growth either between the insulin-alone group
and the methotrexate-alone group or between before and after
treatment in the insulin-alone group.
Treatment
All the patients included in the study received two 21-day courses
of treatment separated by a 7-day interval without treatment be-
tween courses. In group 1, the treatment course was intravenous
human recombinant insulin (0.3 U/kg body weight every other
day) followed 20 min later by a 15-min intravenous infusion of
methotrexate (2.5 mg/m
2
in 50 ml 30% glucose). If symptomatic
hypoglycemia was observed, the 30% glucose solution containing
methotrexate was infused immediately. An oral glucose supplement
was also prescribed to prevent delayed hypoglycemic symptoms. In
group 2, insulin was omitted and methotrexate was administered
intravenously at the same dose and in the same solution (2.5 mg/m
2
in 50 ml 30% glucose) as in group 1. In group 3, methotrexate was
omitted, insulin was administered at the same dose as in group 1,
and 30% glucose solution was also administered intravenously
20 min after insulin or sooner if hypoglycemic symptoms were
evident.
Tumor growth assessment
After 8 weeks (two 3-week courses plus 1 week interval after each
course), the response to treatment was assessed in each patient
using RECIST criteria [13]. The sum of the longest diameter of
measurable target lesions and the number of non-target lesions
were recorded immediately before and after this 8-week period.
Skin nodules and palpable lymph nodes were measured using
calipers. Lung and liver target lesions were measured by a CAT
scan. Responses were confirmed by repeating the assessment
4 weeks after status assignment. Three independent reviewers per-
formed all image measures (Telemedical Organization, North
Miami Beach, Fl.).
The distribution of RECIST status (progressive disease, stable
disease, or remission) in each group was recorded. This distribution
was dependent on treatments that showed statistical significance
according to the Chi-squared test. The data from the RECIST
measurements of the change in tumor size of the patients in each
treatment group, expressed as a percentage of pretreatment
measurements, were compared using Students t-test. Additionally,
increases in tumor size were expressed as a proportion of the initial
value and analyzed by the two-proportion test comparing pairs of
groups: group 3 vs group 1, and group 2 vs group 1. The sample
size was assessed after analysis of the results when the trial was
finished for the 30 patients allocated to the three groups. The above
pairs of groups were analyzed for the proportion of progressive
disease in each. Ten patients in each group was the required sample
size for an 80% chance of rejecting the hypothesis of equal pro-
portions at the 0.05 level of significance when the true proportions
were those shown by the study. Statistical analysis was performed
using StatsDirect software and an independent expert was con-
sulted.
Results
The characteristics of the patients included are shown in
Table 1. The three groups were comparable in the most
relevant prognostic parameters for the clinical condition
studied. Previous treatments were also comparable. The
similar range of sizes of target lesions measured before
treatment was especially significant, allowing the change
in size to be measured as a percentage of initial size.
221
Figure 1 shows the RECIST status assessed und er the
study conditions. Progressive disease was the most fre-
quent response in two of the three groups: in group 2
(treated with methotrexate alone) there were seven
progressive disease and three stable disease, and in
group 3 (treated with insulin alone) there were eight
progressive disease and two stable disease. In group 1
(treated with insulin + methotrexate), stable disease was
the most frequent response (nine stable disease, one
progressive disease). The distribution of RECIST type
responses (stable disease and progressive disease) was
dependent on the treatments tested, and was statistically
significant (P<0.01, Chi-squared test).
Figure 2 shows the means and 95% confidence
intervals (CI) of the percentage increase in tumor size
after treatment in the three groups. Incre ases in tumor
size were significantly lower in patients treated with
insulin + methotrexate than in those treated with
insulin alone and significantly lower than in those trea-
ted with methotrexate alone.
From the same set of measurements, Figs. 1 and 2
show the clinical and biological effects of the treatments,
respectively. Figure 1 indicates that the decrease in tu-
mor growth induced by insulin + methotrexate reached
the level of a clinically confirmed antitumoral response
because more patients in this group achieved stable
disease. Figure 2 shows that insulin + methotrexate
treatment reduced tumor growth. All patients completed
the study. Hypoglycemia was induced in all patients
receiving insulin as part of their protocol. Eight patients
in group 1 and nine patients in group 3 showed no
hypoglycemic symptoms during the 20 min after insulin
injection; they showed a mean blood glucose level of
456 mg/dl (range 376–520 mg/dl). Two patients in group
Table 1 Clinical characteristics
of the 30 women with
metastatic breast cancer (M1)
included in the three treatment
groups
Group 1
(insulin + methotrexate)
Group 2
(methotrexate)
Group 3
(insulin)
No. of patients 10 10 10
Age range (years) 42–64 44–68 39–69
<50 years 4 3 4
Estrogen receptor-positive 7 7 6
Progesterone receptor-positive 7 5 7
Measurable M1
Lung 6 4 4
Liver 1 2 2
Skin 2 2 3
Lymph nodes 1 2 1
Range of initial (pretreatment)
RECIST sum of target
measures (mm)
57–65 59–64 56–66
Fig. 1 Post-treatment RECIST status of measurable target lesions.
After the respective treatment, the change in the measurable lesions
selected as targets in each patient was evaluated and the status of
therapeutic response, defined in terms of the RECIST criteria, was
recorded. Under the conditions of this study, two response statuses
were recorded: stable disease (less than 20% increase or less than
30% decrease in the sum of largest diameters of targets) and
progressive disease (more than 20% increase in the sum of
diameters). Stable disease, the best response obtained, was more
frequent in the group treated with insulin + methotrexate (nine of
ten) than in methotrexate-treated group (three of ten) or insulin-
treated group (two of ten). The distribution of RECIST type
responses (stable disease or progressive disease) was dependent on
the treatments tested and statistically significant (P<0.01, Chi-
squared test)
Fig. 2 Increase in size of measurable target lesions (RECIST
assessment). After each treatment, the change in the measurable
lesions selected as targets in each patient was evaluated in terms of
the RECIST criteria and expressed as a percentage of the measured
pretreatment size. For each treatment group, the mean±SD and
95% CI for the values of this response were calculated: group 1
(insulin + methotrexate) 13.51±3.01% (95% CI 11.35–15.67%);
group 2 (methotrexate) 20.21±2.27% (95% CI 18.58–21.84%);
group 3 (insulin) 21.04±2.17% (95% CI 19.49–22.59%). The
increase in size of lesions in group 1 (insulin + methotrexate) was
significantly lower (Students t-test) than the increase in size in
group 2 (methotrexate) (P<0.001) and group 3 (insulin)
(P<0.001). Group 2 showed no significant difference from group
3(P=0.41)
222
1 and one patient in group 3 showed hypoglycemic
symptoms within 20 min of insulin injection (13, 16 and
19 min), but recovered immediately after starting the
glucose infusion. There was no evidence of any harmful
sequelae attributable to the hypoglycemia induced.
Table 2 shows the toxicities associated with antitu-
moral chemotherapy (according to WHO criteria)
recorded in this study.
Discussion
The methotrexate dose used in this study was chosen
because a similar dose of methotrexate had been used
previously in patients receiving low-dose combined
chemotherapy potentiated with insulin [2]. In addition,
the cumulative monthly dose was no higher than the
monthly dose used in the well-known standard protocol
of methotrexate + fluorouracil + cyclophosphamide
(CMF). Indeed, each individual methotrexate injection
(2.5 mg/m
2
) was less than the dose usually co nsidered
optimal in non-potentiated protocols but is within the
presumed range of effective dose for a potentiation
similar to the one observed in vitro. The results of this
study confirmed the expected safety of the selected
methotrexate dose. The toxicities in the methotrexate-
alone group were not relevant (WHO grades 1/2) and
they were even lower when methotrexate was associated
with insulin, only producing a grade 1 mucositis. In this
study, methotrexate at this safe low dose did not have an
antitumoral effect when used alone (group 2), but it did
produce a significant antitumoral effect when adminis-
tered after insulin (group 1). The term antitumoral is
used here as a description of the clinical effect of a
reduction in the proportion of patients showing pro-
gressive disease.
Therefore, as rep orted previously, our results support
the hypothesis that insulin can potentiate the antitu-
moral effect of methotrexate [2] and confirm in vivo
previously reported in vitro results [10]. Our results also
show insulin potentiation of methotrexate in this con-
dition, where insulin alone did not promote an increase
in tumor growth (group 3). This effect is in agreement
with previous results from in vitro models where insulin
enhancement of cytotoxicity was not a direct conse-
quence of an insulin-dependent increase in the growth
rate of tumor cells [1, 10]. The same in vitro models do
not allow an explanation of the insulin potentiation of
methotrexate in terms of the known effects of insulin
treatment upon the specific metab olism of methotrexate
which include a decrease in intracellular pH induced by
glucose metabolism and tight binding of the drug to its
target, dihydrofolate reductase. Insulin pote ntiation of
other antitumoral drugs has been reported [9].
If we discount the promotion of tumor cell growth
and the interaction with the specific target as the
mechanism of potentiation of methotrexate by insulin,
we can hypothesize that this mechanism could involve
another general insulin-dependent biochemical pathway
as has been previously suggested to explain the in vitro
potentiation of methotrexate by insulin [1]: protein
synthesis in tumor cells is one of the biochemical path-
ways activated by insulin [8]. Most chemotherapy drugs
that have been tested using insulin to increase cytotox-
icity are known modifiers of protein stru cture that act at
the genetic or epigenetic level [12]. High levels of mu-
tated or epigenetically modified proteins could be
responsible for the cytotoxic mechanism elicited by the
insulin-dependent increase in protein synthesis associ-
ated with chemotherapy drugs. The relative selectivity of
this mechanism of action for insulin + methotrexate in
malignant cells is attributed to the agonism of insulin
and insulin-like receptors in tumor cells. Certainly, the
response to insulin is more intense in most tested cancer
cells than in most normal cells. This is probably because
cancer cells are richer in receptors for insulin-like growth
factors that are cross-stimulated by insulin [4].
Conclusion
The in vitro potentiation of methotrexate cytotoxicity by
insulin in human breast cancer cell lines was previously
known. We report the results of a randomized, co n-
trolled trial that confirmed, at the clinical level, the
potentiation by insulin of the antitumoral effect of
methotrexate in women with advanced breast cancer.
The term antitumoral is used as a description of the
clinical effect of a reduction in the proportion of patients
with progressive disease. Under the conditions of this
study, the dose of insulin used did not increase tumor
growth. Therefore, we suggest that, as has been reported
Table 2 Maximum recorded WHO toxicity grade in the patients
included in the trial comparing insulin + methotrexate (group 1),
methotrexate (group 2) and insulin (group 3). The numbers of
patients with each toxicity grade (0 to 4) in the three groups are
shown. No other toxicities referred to in the WHO criteria were
recorded
Toxicity Grade
01234
Erythrocytes
Group 1 10 0 0 0 0
Group 2 8 2 0 0 0
Group 3 10 0 0 0 0
Leukocytes
Group 1 10 0 0 0 0
Group 2 6 3 1 0 0
Group 3 10 0 0 0 0
Platelets
Group 1 10 0 0 0 0
Group 2 9 1 0 0 0
Group 3 10 0 0 0 0
Mucositis
Group 1 8 2 0 0 0
Group 2 4 3 3 0 0
Group 3 10 0 0 0 0
223
in vitro, methotrexate potentiation by insulin was not a
direct consequence of the expansion of the tumor cycling
cell population but a consequence of some of the bio-
chemical events that are simultaneously activated. The
enhancement of methotrexate uptake by tumor cells
and/or the promotion of protein synthesis in a muta-
genic intracellular environme nt are hypothesized to be
mechanisms of potentiation. It is known that both
events are promoted by insulin acting as a cross-agonist
of the highly expressed receptors for insulin-like growth
factors in breast cancer cells.
These mechanisms, which are shared with other pri-
mary tumor cells and with other chemotherapeutic
agents suggest that it would be worthwhile to pursue
further study of these phenomena in other tumors and
with other chemotherapeutic agents.
References
1. Alabaster A, Vonderhaar B, Shafie S (1981) Metabolic modi-
fication by insulin enhances methotrexate cytotoxicity in
MCF-7 human breast cancer cells. Eur J Cancer Clin Oncol
17:1223–1228
2. Ayre SG, Perez Garcia y Bellon D, Perez Garcia D Jr (1990)
Neoadjuvant low-dose chemotherapy with Insulin in breast
carcinomas. Eur J Cancer 26:1262–1263
3. Cardoso F, Di LA, Lohrisch C, Bernard C, Ferreira F, Piccart
M (2002) Second and subsequent lines of chemotherapy for
metastatic breast cancer: what did we learn in the last two
decades. Ann Oncol 13:197–207
4. Cullen KJ, Yee D, Sly WS, Perdue J, Hampton B, Lippman
ME, Rosen N (1990) Insulin-like growth factor receptor
expression and function in human breast cancer. Cancer Res
50:48–53
5. Gross GE, Boldt DH, Osborne CK (1984) Perturbation by
insulin of human breast cancer cell kinetics. Cancer Res
44:3570–3575
6. Kath R, Schiel R, Muller UA, Hoffken K (2000) Malignancies
in patients with insulin-treated diabetes mellitus. J Cancer Res
Clin Oncol 126:412–417
7. Mink PJ, Shahar E, Rosamond WD, Alberg AJ, Folsom AR
(2002) Serum insulin and glucose levels and breast cancer
incidence: the atherosclerosis risk in communities study. Am
J Epidemiol 156:349–352
8. Osborne CK, Bolan G, Monaco ME, Lippman ME (1976)
Hormone responsive human breast cancer in long-term tissue
culture: effect of insulin. Proc Natl Acad Sci U S A 73:4536–
4540
9. Oster JB, Creasey WA (1981) Enhancement of cellular uptake
of ellipticine by insulin preincubation. Eur J Cancer Clin Oncol
17:1097–1103
10. Schilsky RL, Bailey BD, Chabner BA (1981) Characteristics of
membrane transport of methotrexate by cultured human breast
cancer cells. Biochem Pharmacol 30:1537–1542
11. Shackney SE, McCormack GW, Cocheral GJ (1978) Growth
rate patterns of solid tumors and their relation to responsive-
ness to therapy. Ann Intern Med 89:107–121
12. Silva JM, Garcia JM, Dominguez G, Silva J, Rodriguez R,
Portero JL, Corbacho C, Provencio M, Espana P, Bonilla F
Silva JM, Garcia JM, Dominguez G, et al (2000) DNA
damage after chemotherapy correlates with tumor response
and survival in small cell lung cancer patients. Mutat Res
456:65–71
13. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan
RS, Rubinstein L, Verweij J, Van Glabbeke M, van
Oosterom AT, Christian MC, Gwyther SG (2000) New
guidelines to evaluate the response to treatment in solid tu-
mors. European Organization for Research and Treatment of
Cancer, National Cancer Institute of the United States,
National Cancer Institute of Canada. J Natl Cancer Inst
92:205–216
224
... Our findings were consistent with the previous literature. 12,13 In our study, 40 µg/mL of insulin alone for 8 h of incubation had no significant effect on cell growth (Figure 1 ...
... Although the role of insulin in cancer therapy is not fully understood, there are studies reporting its novel application. In a clinical trial conducted by Lasalvia-Prisco et al., 12 it was reported that combination of methotrexate and insulin produced a significantly better clinical outcome in patients with multidrug-resistant metastatic breast cancer, compared to patients treated with methotrexate or insulin alone. In vitro studies [12][13][14][15] found that activity of various chemotherapeutic agents, such as paclitaxel, 5FU, cisplatin, or methotrexate, can be considerably enhanced in the presence of insulin. ...
... In a clinical trial conducted by Lasalvia-Prisco et al., 12 it was reported that combination of methotrexate and insulin produced a significantly better clinical outcome in patients with multidrug-resistant metastatic breast cancer, compared to patients treated with methotrexate or insulin alone. In vitro studies [12][13][14][15] found that activity of various chemotherapeutic agents, such as paclitaxel, 5FU, cisplatin, or methotrexate, can be considerably enhanced in the presence of insulin. It was also reported in a small-scale clinical trial that insulin improves the outcome of hormone therapy in castration-resistant prostate cancer. ...
Article
Full-text available
The study was designed to evaluate the potential use of insulin for cancer-specific treatment. Insulin-induced sensitivity of MCF-7 breast cancer cells to chemotherapeutic agents 5-fluorouracil and cyclophosphamide was evaluated. To investigate and establish the possible mechanisms of this phenomenon, we assessed cell proliferation, induction of apoptosis, activation of apoptotic and autophagic pathways, expression of glucose transporters 1 and 3, formation of reactive oxygen species, and wound-healing assay. Additionally, we reviewed the literature regarding theuse of insulin in cancer-specific treatment. We found that insulin increases the cytotoxic effect of 5-fluorouracil and cyclophosphamide in vitro up to two-fold. The effect was linked to enhancement of apoptosis, activation of apoptotic and autophagic pathways, and overexpression of glucose transporters 1 and 3 as well as inhibition of cell proliferation and motility. We propose a model for insulin-induced sensitization process. Insulin acts as a sensitizer of cancer cells to cytotoxic therapy through various mechanisms opening a possibility for metronomic insulin-based treatments.
... The conflicting results regarding the effect of insulin on the PI3K pathway may be explained due to the relatively short duration of action. Moreover, the concentration of insulin used in the in vitro experiment was higher than circulating concentrations observed in a clinical setting or concentrations used in the previous studies [9][10][11][12][13][14][15][16] . Higher doses of insulin are known to activate IGF1R-related signaling and affect the propensity for invasion and metastasis. ...
... GRB2 signaling acts as a controlling stage and is crucial for cell cycle advancement and cell motility, and therefore, more intricate processes such as morphogenesis, angiogenesis, and vasculogenesis 28 . Research shows that many types of malignancies, such as breast cancer, are characterized by upregulated GRB2-MAPK pathway, especially since GRB2 is coded in a chromosome region duplicated in many types of neoplasms 15,29,30 . Upregulated GRB2 is also correlated with the likelihood of metastasis in colon cancer 31 . ...
Article
Full-text available
The present state of cancer chemotherapy is unsatisfactory. New anticancer drugs that marginally improve the survival of patients continue to be developed at an unsustainably high cost. The study aimed to elucidate the effects of insulin (INS), an inexpensive drug with a convincing safety profile, on the susceptibility of colon cancer to chemotherapeutic agents: 5-fluorouracil (FU), oxaliplatin (OXA), irinotecan (IRI), cyclophosphamide (CPA) and docetaxel (DOC). To examine the effects of insulin on cell viability and apoptosis, we performed an in vitro analysis on colon cancer cell lines Caco-2 and SW480. To verify the results, we performed in vivo analysis on mice bearing MC38 colon tumors. To assess the underlying mechanism of the therapy, we examined the mRNA expression of pathways related to the signaling downstream of insulin receptors (INSR). Moreover, we performed Western blotting to confirm expression patterns derived from the genetic analysis. For the quantification of circulating tumor cells in the peripheral blood, we used the maintrac method. The results of our study show that insulin-pretreated colon cancer cells are significantly more susceptible to commonly used chemotherapeutics. The apoptosis ratio was also enhanced when INS was administered complementary to the examined drugs. The in vivo study showed that the combination of INS and FU resulted in significant inhibition of tumor growth and reduction of the number of circulating tumor cells. This combination caused a significant downregulation of the key signaling substrates downstream of INSR. The results indicate that the downregulation of PIK3CA (phosphatidylinositol 3-kinase catalytic subunit alpha), which plays a critical role in cell signaling and GRB2 (growth factor receptor-bound protein 2), a regulator of cell proliferation and differentiation may be responsible for the sensitizing effect of INS. These findings were confirmed at protein levels by Western blotting. In conclusion, these results suggest that INS might be potentially applied to clinical use to enhance the therapeutic effectiveness of chemotherapeutic drugs. The findings may become a platform for the future development of new and inexpensive strategies for the clinical chemotherapy of tumors.
... Its impact on cellular uptake of many compounds including glucose by facilitated diffusion has been documented [20]. The use of insulin for cancer-specific treatment has been tested in several studies [21][22][23][24][25]. ...
... Due to its complex influence on malignant cell metabolism, insulin has been exploited as a potential sensitizing agent in cancer therapy. Several in vitro and clinical studies have found that the inhibitory effect of various cytotoxic agents can be enhanced in the presence of insulin [21][22][23][24][25]. ...
Article
Full-text available
The rationale for the implementation of novel therapies should be based on hallmarks of cancer. Two novel compounds labelled as thioglycoside A and B were designed and evaluated on breast and colon cancer cell lines. We assessed their cytotoxic effect after sensitizing cancer cells with insulin. In order to explore the underlying mechanisms, we performed tests to assess cell migration and motility, apoptosis, expression of glucose transporter 1 and proapoptotic proteins. Both compounds proved to have an antitumor effect which was significantly enhanced in combination with insulin. Linking glucose and anticancer agent presents an approach that exploits the Warburg effect. Targeting dysfunctional glycometabolism and increased glucose absorption is emerging as a promising anticancer strategy.
... In a clinical trial involving multidrug resistant metastatic BC (resistant to fluorouracil + Adriamycin/doxorubicin + cyclophosphamide), a combination of insulin administration enhanced the antitumoral response to methotrexate. It significantly lowered the median increase in tumor size when compared to those who were administered with either insulin or methotrexate alone [220]. However, the mechanism of insulin-mediated potentiation of chemosensitivity is not clear yet. ...
Article
Full-text available
Breast cancer (BC) is the most prevalent cancer in women. BC is heterogeneous, with distinct phenotypical and morphological characteristics. These are based on their gene expression profiles, which divide BC into different subtypes, among which the triple-negative breast cancer (TNBC) subtype is the most aggressive one. The growing interest in tumor metabolism emphasizes the role of altered glucose metabolism in driving cancer progression, response to cancer treatment, and its distinct role in therapy resistance. Alterations in glucose metabolism are characterized by increased uptake of glucose, hyperactivated glycolysis, decreased oxidative phosphorylation (OXPHOS) component, and the accumulation of lactate. These deviations are attributed to the upregulation of key glycolytic enzymes and transporters of the glucose metabolic pathway. Key glycolytic enzymes such as hexokinase, lactate dehydrogenase, and enolase are upregulated, thereby conferring resistance towards drugs such as cisplatin, paclitaxel, tamoxifen, and doxorubicin. Besides, drug efflux and detoxification are two energy-dependent mechanisms contributing to resistance. The emergence of resistance to chemotherapy can occur at an early or later stage of the treatment, thus limiting the success and outcome of the therapy. Therefore, understanding the aberrant glucose metabolism in tumors and its link in conferring therapy resistance is essential. Using combinatory treatment with metabolic inhibitors, for example, 2-deoxy-D-glucose (2-DG) and metformin, showed promising results in countering therapy resistance. Newer drug designs such as drugs conjugated to sugars or peptides that utilize the enhanced expression of tumor cell glucose transporters offer selective and efficient drug delivery to cancer cells with less toxicity to healthy cells. Last but not least, naturally occurring compounds of plants defined as phytochemicals manifest a promising approach for the eradication of cancer cells via suppression of essential enzymes or other compartments associated with glycolysis. Their benefits for human health open new opportunities in therapeutic intervention, either alone or in combination with chemotherapeutic drugs. Importantly, phytochemicals as efficacious instruments of anticancer therapy can suppress events leading to chemoresistance of cancer cells. Here, we review the current knowledge of altered glucose metabolism in contributing to resistance to classical anticancer drugs in BC treatment and various ways to target the aberrant metabolism that will serve as a promising strategy for chemosensitizing tumors and overcoming resistance in BC.
... Giving insulin just prior to the infusion of low dose chemotherapeutic agents, usually in a combination designed to intervene at several sites of the cell cycle, causes much less side effects and can hold the cancer at bay while buying time to reconstitute an effective immune response or integrate another therapeutic strategy. IPT has been in development and clinical use since the 1930s and has been used to help treat other chronic diseases as well [40][41][42][43][44][45][46][47][48]. ...
Article
Full-text available
Over the years, the treatment of patients with cancer has varied widely as much because of recent advancements in science and medicine as the philosophies that belie their use. This paper briefly describes many of the prevailing approaches in use today with an attempt to offer some perspective of how to apply these disparate methodologies so that they may be more effectively integrated, resulting in consistently better clinical responses.
... 52 It follows that the antitumor activity of insulin may allow this hormone to be used in combination with chemotherapy to treat cancer. [53][54][55] Conclusion l-Glutamine supplementation of diet for 10 days in a cancer cachexia animal model promoted increased PEPCK expression in the jejunum that was associated with a significant elevation of glycemia and insulinemia, suggesting a beneficial effect on energy balance. An indirect effect on insulin secretion by l-glutamine is likely to prevent catabolism, thus reducing the harmful effects of cachexia. ...
Article
We evaluated the effects of supplementation with oral l-glutamine in Walker-256 tumor–bearing rats. A total of 32 male Wistar rats aged 54 days were randomly divided into four groups: rats without Walker-256 tumor, that is, control rats (C group); control rats supplemented with l-glutamine (CG group); Walker-256 tumor rats without l-glutamine supplementation (WT group); and WT rats supplemented with l-glutamine (WTG group). l-Glutamine was incorporated into standard food at a proportion of 2 g/100 g (2%). After 10 days of the experimental period, the jejunum and duodenum were removed and processed. Protein expression levels of key enzymes of gluconeogenesis, that is, phosphoenolpyruvate carboxykinase and glucose-6-phosphatase, were analyzed by western blot and immunohistochemical techniques. In addition, plasma corticosterone, glucose, insulin, and urea levels were evaluated. The WTG group showed significantly increased plasma glucose and insulin levels (p < 0.05); however, plasma corticosterone and urea remained unchanged. Moreover, the WTG group showed increased immunoreactive staining for jejunal phosphoenolpyruvate carboxykinase and increased expression of duodenal glucose-6-phosphatase. Furthermore, the WTG group presented with less intense cancer cachexia and slower tumor growth. These results could be attributed, at least partly, to increased intestinal gluconeogenesis and insulinemia, and better glycemia maintenance during fasting in Walker-256 tumor rats on a diet supplemented with l-glutamine.
... In previous IPT clinical trials [110,111], cancer patients were administered insulin 0.3 to 0.4 U per kg body weight intravenously, followed by chemotherapy at lower doses than usual, starting 20 min after insulin administration or sooner if symptoms of hypoglycemia were observed. In another case study [112], a woman with breast cancer was treated with IPT. ...
Article
Full-text available
Introducing a new drug to the market is a time-consuming process, is complex, and involves consumption of a lot of resources. Therefore, discovering new uses for the old drugs (i.e. drug repurposing) benefits the patients by providing them time-tested drugs. With developments in insulin therapy still happening, it is worth keeping up to date on trends in the use of this powerful glucose-lowering agent. The aim of this article is to explore the potential non-diabetic clinical applications of insulin. Literature survey was carried out through the various scientific journals publishing experimental and clinical research papers regarding the diverse applications of insulin other than in diabetes mellitus. These applications include both therapeutic as well as diagnostic uses of insulin. The relevant information collected from these publications was paraphrased in the present paper. On studying the literature, the non-diabetic uses of insulin include the following: wound healing, parenteral nutrition, antiaging, body building, cardioprotection in acute coronary syndromes, insulin tolerance test to test the hypothalamo-pituitary-adrenal axis functioning, cell culture, cancer treatment, organ preservation, and management of septic shock, calcium channel, β blocker overdose and other critical illnesses in intensive care units. This review attempts to survey some interesting new applications of insulin other than in diabetes mellitus.
Article
Full-text available
The global burden of cancer incidence, deaths and economic costs is steadily increasing since several decades. Despite a massive allocation of research funds since the 1970s, no significant (in terms of years) improvements of survival times have been achieved for most cancer types. In this article, I argue that the failure to effectively prevent and treat cancer is partly owing to the gene-centric paradigm of the somatic mutation theory of carcinogenesis. I outline and provide evidence for a new transdisciplinary evolutionary theory of carcinogenesis according to which cancer is a phylogenetic reversal towards unicellular lifeforms triggered by the breakdown of essential cooperative interactions on important levels of human organization. These levels include the genetic, cellular, tissue and psychosocial-spiritual level of human existence. The new theory considers the emergence of eukaryotes and metazoans and e of particular importance e human evolution and in this way explains why cooperation on these different levels is so essential to maintain holistic health. It is argued that the interaction between human's slow natural evolution and the fast cultural evolution, especially during the current Anthropocene epoch, plays an important role in making individuals susceptible towards carcinogenesis. The implications of this insight and the theory of cancer as a phylogenetic reversal are discussed with respect to prevention and treatment, and concrete practical examples are provided. It is concluded that individuals could substantially reduce their risk of cancer by respecting certain biopsychosocial-spiritual lifestyle factors which are justified by human evolution.
Article
Anthracyclines have been important and effective treatments against a number of cancers since their discovery. However, their use in therapy has been complicated by severe side effects and toxicity that occur during or after treatment, including cardiotoxicity. The mode of action of anthracyclines is complex, with several mechanisms proposed. It is possible that their high toxicity is due to the large set of processes involved in anthracycline action. The development of resistance is a major barrier to successful treatment when using anthracyclines. This resistance is based on a series of mechanisms that have been studied and addressed in recent years. This work provides an overview of the anthracyclines used in cancer therapy. It discusses their mechanisms of activity, toxicity, and chemoresistance, as well as the approaches used to improve their activity, decrease their toxicity, and overcome resistance.
Article
The present work reports on the investigation of the possible influence of insulin on the photodynamic effect of the photosensitizer radachlorin on four cell lines: HepG2, HT-29, BJ and BALB/3T3. For a quantitative comparison, the insulin effect on the standard chemotherapy drug methotrexate is also measured. The results show that the EC(Formula presented.) value for methotrexate is 40% higher than when combined with insulin; and for the lowest methotrexate concentration used, the difference exceeds 120%. For the case of the photodynamic effect, it is shown that insulin, when applied in combination with radachlorin, improves the cell permeability for the photosensitizer, thus increasing its concentration in the cells. The cytotoxic effect of photodynamic therapy with radachlorin as the photosensitizer has been quantitatively compared. The quantitative comparison of the photodynamic effect in the cases of using the photosensitizer radachlorin both alone and in combination with insulin, shows that the addition of insulin does not have an effect on the model of normal cell lines used; while for the HepG2 tumor cells, a two-fold increase in the EC(Formula presented.) value is measured.
Article
Full-text available
The insulin-like growth factors IGF-I and Idi -li are potent mitogens for several breast tumor cell lines in culture. Additionally, both IGF-I and IGF-II mRNAs are easily detected in the majority of breast tumor specimens examined, while no breast cancer epithelial cell lines we have studied express authentic IGF-I mRNA, and few lines express IGF-II iiiRNA. Although receptors for insulin, IGF-I, and IGF-II have been described, there is significant cross-reactivity between the various recep tors and ligands in the insulin/insulin-like growth factor family, and it is not clear which receptor or receptors are responsible for the biological effects of these growth factors in this system. Using an RNase protection assay, we examined breast tumor speci mens and breast cancer epithelial cell lines for expression of mRNA encoding the type I and type II IGF receptors as well as the insulin receptor. Virtually all of the specimens examined expressed mRNA for all three receptors. We then examined estrogen-dependent MCF-7 cells for the mitogenic effects of IGF-I and II in the presence of antibodies to both the type I and type II receptors. aIR-3, a monoclonal antibody which blocks the type I receptor, abolished the mitogenic effects of both IGF-I and IGF-II. It did not, however, block the mitogenic effects of insulin. We conclude that type I and type II IGF receptors are ubiqui tously expressed in breast cancer, and our experiments with MCF-7 cells suggest the mitogenic effects of both IGF-I and IGF-II are mediated via the type I IGF receptor.
Article
Full-text available
Anticancer cytotoxic agents go through a process by which their antitumor activity—on the basis of the amount of tumor shrinkage they could generate—has been investigated. In the late 1970s, the International Union Against Cancer and the World Health Organization introduced specific criteria for the codification of tumor response evaluation. In 1994, several organizations involved in clinical research combined forces to tackle the review of these criteria on the basis of the experience and knowledge acquired since then. After several years of intensive discussions, a new set of guidelines is ready that will supersede the former criteria. In parallel to this initiative, one of the participating groups developed a model by which response rates could be derived from unidimensional measurement of tumor lesions instead of the usual bidimensional approach. This new concept has been largely validated by the Response Evaluation Criteria in Solid Tumors Group and integrated into the present guidelines. This special article also provides some philosophic background to clarify the various purposes of response evaluation. It proposes a model by which a combined assessment of all existing lesions, characterized by target lesions (to be measured) and nontarget lesions, is used to extrapolate an overall response to treatment. Methods of assessing tumor lesions are better codified, briefly within the guidelines and in more detail in Appendix I. All other aspects of response evaluation have been discussed, reviewed, and amended whenever appropriate.
Article
Insulin, which activates and modifies metabolic pathways in MCF-1 human breast cancer cells, is shown to increase the cytotoxic effect of methotrexate up to ten thousand-fold in vitro. This enhanced Cytotoxicity is not due to an increased bound intracellular drug level, an increased growth rate or an increase in S phase cells, but may involve the modification or activation of biochemical pathways associated with cell growth, even in cells not undergoing DNA synthesis. This observation supports the hypothesis that tumor cell sensitivity to chemotherapy could be increased by using agents that can activate the biochemical or metabolic pathways that determine the cytotoxic process.
Article
The clinical doubling times of human solid tumors are reviewed. Methods are also described for estimating subclinical tumor doubling times, and these methods are applied to Wilms' tumor, choriocarcinoma, Burkitt's tumor, and breast cancer. Rapidly growing tumors are often responsive to therapy, and such complete responses are often quite durable. Slowly growing tumors respond less favorably to therapy, and responses are generally not durable. Rapidly growing tumors in advanced stages are similar in their response behavior to the slowly growing tumors. A strategy based on cell kinetics principles and tumor doubling time data is proposed for improving therapeutic results in responsive tumors. Data on clinical and subclinical tumor doubling times are used to distinguish early recurrences from late recurrences in a given tumor type. Early recurrences call for intensification of induction therapy, whereas late recurrences call for prolongation of consolidative therapy. This strategy may also apply in adjuvant therapy of slowly growing tumors.
Article
The effects of estrone, estradiol, and estriol on MCF-7 human breast cancer are compared. In this estrogen-responsive cell line, all three estrogens are capable of inducing equivalent stimulation of amino acid and nucleoside incorporation. Estriol is capable of partially overcoming antiestrogen inhibition with Tamoxifen (lCl 46474), even when antiestrogen is present in 1000-fold excess. Antiestrogen effects are completely overcome by 100-fold less estriol. Studies of metabolism of estrogens by MCF-7 cells revealed no conversion of estriol to either estrone or estradiol. All three steroids bind to a high-affinity estrogen receptor found in these cells. The apparent dissociation constant is lower for estradiol than for estrone and estriol, but all three bind to an equal number of sites when saturating concentrations are used. Tritiated estrogens used in binding studies were shown to be radiochemically pure. We conclude that estriol can bind to estrogen receptor and stimulate human breast cancer in tissue culture. Our data do not support an antiestrogenic role for estriol in human breast cancer.
Article
The mechanisms of steroid and peptide hormone action in human breast cancer are poorly understood. We have previously characterized a cell line of human breast cancer in long-term tissue culture that possesses various steroid hormone receptors and responses, providing a model for the study of steroid hormone action. The present studies describe a human breast cancer in vitro that responds to physiologie concentrations of insulin with an increased rate of macromolecular synthesis and growth. Thymidine and uridine incorporation in cells in serum-free medium are stimulated by 10(-11) M insulin and are maximal with 10(-8) M. Leucine incorporation is stimulated by 5 X 10(-11) M insulin and is maximal with 10(-9) M. Significant stimulation of uridine and leucine incorporation is evident by 3 hr and maximal by 10 hr. A 10-hr lag period exists for insulin stimulation of thymidine incorporation, which is maximal form 14 to 24 hr. The effect of 10(-8) M insulin on macromolecular synthesis is accompanied by a 69% increase above controls in the number of cells after 24 hr. The effect on macromolecular synthesis is observed in glucose-free medium. Insulin's effect on protein synthesis is not blocked by inhibition of RNA synthesis with actinomycin D. Glucocorticoids partially inhibit the action of insulin in these cells. This system provides a model for studying insulin action, and suggests that some human breast cancer may show growth regulation by insulin.
Article
The growth of cultured human breast cancer cells is sensitive to physiological concentrations of insulin suggesting that it may regulate breast cancer growth in vivo. The mechanisms for the growth effects of insulin are poorly defined. In the present study, we examine the effects of insulin on the cell cycle kinetics of asynchronous MCF-7 human breast cancer cells growing in serum-free medium. When the [3H]thymidine labeling index is used to estimate the S-phase fraction, insulin added to asynchronously growing cells results in a time-dependent increase in the proportion of cells engaged in DNA synthesis. Computer analysis of DNA histograms obtained by flow cytometry of mithramycin-stained cells also shows a time-dependent progression of cells into and through the S-phase compartment. Sixteen hr after adding insulin to asynchronous cells, 66% of cells are in S-phase compared to 37% in controls. The effect of insulin on the cell cycle progression of MCF-7 cells is also dose dependent. Stimulation is observed with physiological insulin concentrations of 0.1 to 1.0 nM; maximal effects are observed with 1.0 to 10 nM insulin. Various insulin analogues enhance the progression of cells into S phase in proportion to their ability to bind to the insulin receptor in MCF-7 cells (porcine ≥ chicken > guinea pig > deoctapeptide insulin), while unrelated peptide hormones have no effect on the cell cycle kinetics. Cell cycle analysis after the addition of colchicine to prevent mitosis and the reentry of cells into G1 demonstrates a shortened G1 in response to insulin. Continuous [3H]thymidine-labeling studies after the addition of colchicine suggest that the growth fraction is about 88% with or without insulin. In summary, insulin causes a marked perturbation of the cell cycle kinetics of MCF-7 human breast cancer cells by facilitating the transit of cells through G1. The data also suggest that this effect is mediated via the insulin receptor.
Article
Methotrexate, a folic acid analogue, enters cells using a high-affinity carrier system that is shared with naturally occurring reduced folates. Methotrexate transport by MCF-7 cells, a hormonally responsive line of human breast cancer cells, exhibited a high-affinity carrier system that displayed Michaelis-Menten kinetics [Km = 8.22 ± 0.62 μM; Vmax = 12.22 ± 2.8 nmoles · min−1·(g cell protein)−1] was competitively inhibited by leucovorin and aminopterin, but not by folic acid; and was temperature-sensitive (Q10 = 2.25 ± 0.32). Initial uptake rates were not affected by ouabain or sodium azide, but efflux of intracellular drug was inhibited markedly by sodium azide, suggesting the presence of an energy-dependent active efflux mechanism. At extracellular methotrexate concentrations in excess of 10 μM, a second, low-affinity uptake component could be identified that may represent a lower affinity membrane carrier or passive diffusion. Examination of hormonal influences on methotrexate transport revealed that growth of MCF-7 cells in serum-free medium induced a significant increase in the transport Km value (15.93 ± 1.6 μM) compared to the Km of 8.22 μM for cells grown in fetal calf serum. This change in affinity of the transport carrier could be reversed by the additon of insulin, but not of estradiol, to the culture medium. Methotrexate transport by human breast-cancer cells displayed characteristics that were similar to those of transport reported for human leukemia cells but that have not been documented previously for cells derived from a human solid tumor. In addition, the drug transport carrier was subject to modulation by insulin but not by estrogen.