ArticlePDF AvailableLiterature Review

Insulin Therapy and Cancer

Authors:

Abstract

That diabetes is associated with an increased lifetime risk of developing malignancy is now well established (1,2). For some specific malignancies, explanation of that increased risk is possible, though usually unproven. For example, the marked increase in pancreatic cancer incidence in type 2 diabetes may or may not be due to misattribution of type of diabetes where the reality is secondary diabetes. Also markedly increased is hepatic malignancy—something that may be secondary to hepatic steatosis or, rather, hepatosteatitis and subsequent fibrotic/regenerative disease. Lesser increases in large-bowel malignancy are also found in association with obesity and modern low-residue high-fat diets, and both obesity and such diets are associated with diabetes (1,3). In contrast, it is notable that high-concentration insulin given repeatedly into the same injection site for many decades has not generated even one report of a subcutaneous sarcoma in >90 years in tens of millions of people. Type 2 diabetes and other types of diabetes, once vascular damage is occurring, are inflammatory states, which may provide an environment for oncogenesis of some malignancies. However, the inflammatory state might also might provide enhancement of immune cancer surveillance mechanisms. The reduction of prostate cancer incidence is potentially explainable in the context of relative reduction in circulating testosterone levels in men with type 2 diabetes. A second concern over diabetes and malignancy is regarding glucose-lowering therapies. However, only in the case of PPAR-αγ agonists is there reasonably strong data once information from preclinical studies, randomized controlled trials (RCTs), and observational studies is taken together (4–6). For insulin, the data are more complex to assess. Preclinical animal data of concern are limited to an analog with unusual receptor binding properties. However, mechanistic data do suggest mechanisms by which increased malignancy risk could operate, and this has raised concerns …
Insulin Therapy and Cancer
PHILIP HOME, DM, DPHIL
T
hat diabetes is associated with an
increased lifetime risk of developing
malignancy is now well establish ed
(1,2). For some specic malignancies, ex-
planation of that increa sed risk is possi-
ble, though usually unproven. For
example, the marked increase in pancre-
atic cancer incidence in type 2 diabetes
may or may not be due to misattribution
of type of diabetes where the reality is sec-
ondary diabetes. Also markedly increased
is hepatic malignancydsomething that
may be secondary to hepatic steatosis or,
rather, hepatosteatitis and subsequent
brotic/regenerative disease. Lesser increa-
ses in large-bowel malignancy are also
found in association with obesity and mod-
ern low-r esidue high-fat diets, and both
obesity and such diets are associated with
diabetes (1,3). In contrast, it is notable that
high-conce ntration insul in given repeat-
edly into the same injection site for many
decades has not generated even one report
of a subcutaneous sarcoma in .90 years in
tens of millions of people.
Type 2 diabetes and other types of
diabetes, once vascular dama ge is occ ur-
ring, are inammatory states, which may
provide an environment for oncogenesis
of some malignanc ies. However, the in-
ammatory state might also might pro-
vide enhancement of immune cancer
surveillance mechanisms. The reduction
of prostat e cancer incidence is potentially
expl aina ble in the context o f relative re-
duction in circulating testosterone levels
in m en with type 2 diabetes.
A second concern over diabetes and
malignancy is regarding glucose-low ering
therapies. However, only in the case of
PPAR-ag agonists is there reasonably
strong data once information from pre-
clinical studies, randomized controlled
trials (RCTs), and ob servational studies
is taken together (46). For insulin, the
data are more complex to assess. Preclin-
ical animal data of concern are limited
to an analog with unusual receptor bind-
ing properties. However, mechanistic
data do suggest mechanisms by which in-
creased malignancy r isk could operate,
and this has raised concerns about some
licensed insulin analogs, nota bly insulin
glargine.
This review is meant to be an over-
view of the concerns surrounding insulin,
with the intention of seeking the level of
probability that endogenous or exoge-
nous insulin could be contributing to
malignancy risk in clinical practice and
looking for research pathways to i nform
this fu rther.
METHODSdLiterature searching for
this review was not systematic. Search
terms around insulin, ma lignancy, and
cancer and narrowed for specicareasto
observational studies, randomized con-
trolled trials, insulin therapy, growt h pro-
motion, and insulin analog(ue)s still
revealed very large numbers of sources
in the literature (.2,000,000), from
which the reviewer then selected, with
potential bias, papers of higher quality
or interest and those more often quoted
by other authors and supported by
studies refe rred to from the initially
identied reports.
In han dling the evidence, I at tempt to
take the data from a study in the context
of prior know ledge and asks the question,
‘‘How does this study shift the probability
that such and such an effect is real to the
exte nt that it should help determine rou-
tine clinical practice or promote further
rese arch activity?’’ The limitations of the
available literature restrict the conclu-
sions that can be made, as in any scientic
review.
MECHANISTIC DATAdThe exis-
tence of potential mechanisms by
which a substance (in this case, insulin
or derivatives) can promote malignanc y
does not by itself signi
cantly raise con-
cerns that such a problem will occur.
Indeed, without other evidence, whether
preclinical (animal studies), observa-
tional, or controlled studies, mechanistic
data on how such an effect might occur
provide very little indication of probabil-
ity that it will occur in clinica l practi ce.
This is contra ry to human intuition be-
cause the opposite approach, of attaching
a mechanism to a described adverse event,
provides a good opportunity for avoiding
or ameliorating the event, and accordingly
we instinctively give mechanistic explan-
ations quite a lot or even excessive weight.
As will be seen below, the evidence from
animal and clinical studies for insulin
being a causative factor in human malig-
nancy is weak, so the mechanistic discus-
sions that fol low currently have limited
clinical relevance.
Most of the potential mechanisms
for insulin being a risk factor for cancer
concern growth promotion. Studies
looking for genotoxicity have been un-
successful (7). There is no evidence that
insulin regulates or affects expression of
established oncogenes, includin g those
of the tyrosine kinase family. Growth
promotion c ould affect the lifetime clin-
ical incidence of cancer by allowing pro-
tective cellular and immune surveillance
mechanisms to be overcome. Further-
more, in the short term it might appear
to increase the incidence rate of cancers
by causing thos e already present to be
detected sooner.
Two putative mechanisms may be
identied. Firstly, and attracting much
attention with rega rd of insulin therapy, is
the cross-reactivity between bindi ng for
thei r respective receptors (and subtypes)
of insulin and IGF-1 (8). Insulin is in
any case, and distinct from its glucose-
lowering activity, a n anabolic hormone
with actions on amino acid uptake and
protein synthesis, which is the basis of
its misuse by athletes. Of interest here
is the existence of a growth-promoting
(fetal) insulin receptor (A), which is
cccccccccccccccccccccccccccccccccccccccccccccccc c
From the Institute of Cellular MedicineDiabetes, Newcastle University, Newcastle upon Tyne, U.K.
Corresponding author: Philip Home, philip.home@ncl.ac.uk.
This publication is based on the presentations from the 4th World Congress on Controversies to Consensus in
Diabetes, Obesity and Hypertension (CODHy). The Congress and the publication of this supplement were
made possible in part by unrestricted educational grants from Abbott, AstraZeneca, Boehringer Ingelheim,
Bristol-Myers Squibb, Eli Lilly, Ethicon Endo-Surgery, Janssen, Medtronic, Novo Nordisk, Sano, and
Takeda.
DOI: 10.2337/dcS13-2002
© 2013 by the American Diabetes Association. Readers may use this article as long as the work is properly
cited, the use is educational and not for prot, and the work is not altered. See http://creativecommons.org/
licenses/by-nc-nd/3.0/ for details.
S240 DIABETES CARE, VOLUME 36, SUPPLEMENT 2, AUGUST 2013 care.diabetesjournals.org
DIABETES AND CANCER
sometimes overexpressed in tumors,
notably breast malignancies, and of hy-
brid insulinIGF-1 receptors (9,10). Fur-
thermore, insulin receptor A has higher
binding afnity for IGF-1, though it is still
an order lower than for insulin (11).
Higher concentrations of insulin receptor
substrate 1 can be found in malignant tu-
mors but are reported to be highest in
those with ben ign outcome (12).
Clinically, the property that has
raised concerns for insuli ns has been its
IGF-1toinsulin re ceptor binding afn-
ity. How ever, as insulin is mainly cleared
through its receptor, insu lins with low in-
sulin receptor afnity are present in the
circulation in higher concentration than
native human insulindsomething that
will also enhance IGF-1 receptor binding.
Insulins of the B-terminal diargi nyl series
(i.e., B31-B32 diarginyl insulins) are of
particular conc ern, a s without o ther
amino acid change they have an ~20:1
IGF-1:insulin receptor binding ratio com-
pared with human insulin (8). However,
even at this ratio i t is debatable whether
insulin would contribute s ignicant
growth factor activity compared with the
tissue levels of IGF-1 itself, given that the
latter has much higher afnity for its own
receptor than does insulin and because it
circulates at much higher concentrations
than insulin (11).
Studies of malignancy rates in acro-
meg aly have resulted in uncertainty as to
whether malignancy is increased or not
(13). A particular concern might, how-
ever, arise with breast cancers (other ma-
lignancies are less well studied), some of
which have markedly enhanced expres-
sion of insulin or IGF-1 receptors (14,15).
The second mechanism of insulin
growth promotion is less well documen-
ted but did result in mammary tumors in
susceptible rats. Asp-B10 insulin is an
unusual human insulin analog that h ad
apparently markedly enhanced binding
to hepatocytes i n vitro. However , that
may be an artifact of another rec eptor
property of that insulin, namely, slow
insu lin receptor dissociation rate (16). It
is hypothesized that this propert y might
have bee n the cause of increased signaling
down the mitogen-activated protein ki-
nase pathway (and thus growth prom o-
tion and mitogenesis) rather than the
phosphatidylinositol 3-kinase or CAP
(Cbl-associated protein) pathways in-
volved in metabolic signaling. Studies of
other insulins suggest that this was a fairly
unique property of Asp-B10 insulin; it is
not a problem for any of the other human
insulin analogs in clinical use, such as in-
sulin glargine (17).
PROPERTIES OF INSULIN
GLARGINE dInsulin glargine is an
end-chain diarginyl insulin (see ab ove)
but addit ionally has a Gly-A21 amino acid
substitutionda s ubstitution that reduces
relative IGF-1binding activity (8). A c-
cordingly (and dependent on the cell
model used for testing ) the 320 increase
in binding of the B-chain substitution is
reduced to 36 for insulin glarg ine itself
(8). As noted above, t his difference is
unlikely to be signicant in canc er-
promoting terms, as recognized at the
point of licensing, partic ularly as detailed
receptor studies did not reveal any Asp-
B10 insulinlike continued receptor sig -
naling (17).
Diarginyl insulin is a natural product
of endogenous insulin production and
circulates in al l people with an intact
pancreas, though in very small amounts
relative to insul in. Indeed, the diarginyl
bond is a regular cleav age site in proteins,
and many tissues contain pepti dases to
remove such residues, including subcu-
taneous tissue. T his raises the q uestion as
to whether insulin glargine, during its
long residence time in the subcutaneous
tissues or after rel ease from its insoluble
subcutaneous complexes b ut before ab-
sorption, might be partly or lar gely de-
graded to the A21-Gly human insulin
with low IGF-1 receptor binding. Indeed,
this turns out to be the case, with three
studies reporting that the overwhe lming
circulating species of insulin after sub-
cutaneous glargine injection is its so-
called M1 metabolite, A21-Gly human
insulin ( 1820).
OBSERVATIONAL STUDIES
Methodology and biases
Observational studies with insulin in
people with type 2 diabetes often nd
that it is associated with wors e outcomes
and more deranged metaboli sm (what-
ever outcome is me asured), as indeed is
the case for malignancy (21). In some
studies, it is recognized that for the most
part, sin ce insulin is used late in the ther-
apeutic pathway, it is useful to a djust for
age and duration of diabetes. Unfortu-
nately, even this cannot control f or b iases
introduced by people with other illness
being in closer conta ct with the medical
profession and the risk that in the com-
plex disease environment the cli nical site
(e.g., university hospital) where insulin is
started may be more likely to be using the
latest glucose-lowering technologies (2).
Observational studies should usually
therefore be regarded as hypothesis gen-
erating.
Some studies have suggested insulin to
be associated with lower rates of cancer and
lower rates of progression to an extent that
has been noted by commentators to be
remarkable (22,23). Again, this is likely to
be an association confounded by hidden
factors; people presenting with cancer or
progressing with disseminated disease will
tend to be anorexic, such that if they have
type 2 diabetes the need for therapies will
be reduced.
Biases that can result in confounding
effects in clude drug use indication bias,
prevalent user bias, detecti on bias, and
time-rel ated biases. The potential for in-
dication bias is very large in type 2 di-
abetes owing to the complexity of the
condition and its association with obesity
(and thus dietary change), cardiovascular
risk factors, and inammation . As a sim-
ple example, obesity and hyperglycemia
are associated with malignancy and with
insulin insensit ivitydthus the ea rlier use
of insulin and of insulin in higher doses.
Indication bias might result in people
subject to adverse health outcomes such
as malignancies being more likely to come
into contact with hospital-based care
teams and then more likely to start
diabetes therapies inclu ding insulin ear-
lier than those continuing to be seen in
family practice. This might account for
the detection bias (high rates in early
months falling rapidly th ereafter) seen in
some studies (24). Currie et al. (25) found
that insulin users with cancer were more
likely to have cardiovascular disease at di-
agnosis, conrming that they were not the
same populati on as the comparator non-
insulin users. Lind et al. (26) noted rapid
falls in prost ate cancer incidence over 5
years from abnormally high levels w ith
duration of use of insulin glargine. This
might then even apply to particular ther-
apies if, for example, one group of practi-
tioners were more likely to use insulin
analogs than another using pre do mi-
nantly human insulins. Other issues
surround contraindications and com-
parators; metformin, for example, has
tended to be used less in anyone with
liver, renal, or gut disturbance, so pop-
ulations taking metformin tend to do
better in observational cohorts for
many types of outcomes. However, for
cancer th is w as not conrmed in a meta-
analysis of randomized controlled trials
care. diabe tesjournals.org DIABETES CARE, VOLUME 36, SUPPLEMENT 2, AUGUST 2013 S241
Home
(27). A dditionally, metformin is usually
prescribed early in the course of diabetes
and insulin later, so studies need to ad-
just for duration o f diabetes, which ap-
parently was not done even in a recent
database study (25).
Prevalent user bias can arise in vari-
ous ways and someti mes can be as simple
as poor ascertainment of when a drug is
started in relation to an o utco me (if, for
example, the drug is started in the hospi-
tal but the rst record is later from a
prescription issued in family practice)
(28). The issue of time-related biases in
diabetes therapy studies has been dis-
cussed by Suissa and Azoulay recently
(29).
Outcomes of studies
In the light of the above problems, can
anything useful be concluded from pub-
lished observational studies of cancer (in-
sulin glargine is cons idered separately
below) and insulin use? Even if they are
taken to be hypothesis generating, the
literature is quite contradictory as to asso-
ciations of increased or decreased preva-
lence, incidence, or progression (28).
Given these conicting ndings and the
unresolved methodological issues dis-
cussed above, the best conclusion is per-
haps that the studies as a whole do not
contribute to clinical decision making in
the management of people with diabetes,
although individual studies are useful in
contradicting the ndings of others.
There does seem to be an impro ve-
ment in methodological quality or at-
tempts to address it in the las t 5 y ears.
The China/Hong Kong studies from Yang
and colleagues are helpful in this regard,
but as noted above the ndings from
some of these are surprisingly favorable
with rega rd to insulin therap y to an extent
that is difcult to understand mechanis-
tically (2,22). Furthermore, study of ori-
ental populations with BMI well b elow
30.0 kg/m
2
on average may not help to
answer the underlying question of
whether obese people with unphysio-
logically high (328) endogenous with
or without exogenous insulin exposure
suffer a growth-promoting or other ef-
fect of insulin if they develop susceptible
tumors.Boyleetal.(30)haverecently
published a detailed meta-analysis
of breast cancer in diabetes, conclud-
ingthattheremaybea27%increase
in type 2 diabetes but not in type 1 di-
abetes or gestational diabetes mellitus
where insulin use is universal or more
common.
Insulin glargine
As noted above, the issues surrounding
insulin glargine are not s upported by
mechanistic studies, but once raised the
questions cannot be ignored (31). The
RCT evidence is di scussed separately be-
low. The issue came to prominence with
the publication of four studies in Diabeto-
logia, which taken together suggested no
malignancy association with gl argine, de-
spite being su bject to some of the meth-
odological issues discussed above (32
35). In one study, which suggested an
overall but small benet of association
wit h insu lin glargi ne use, the data were
clearly awed by the study populations
having very different i nsulin dose use,
meaning they were different noncompa-
rable populations for unascertained rea-
sons (32). In t hese circumstances, no
adjustment for the limited range of cova-
riates would help (propensity ana lysis
mighthavegonesomewaytoreducing
biases, using pair matching for all avail-
able characteristics including dos e), and
therefore the authors nding of increased
dose-adjusted risk for glargine has very
little value. In another of the four stud-
ies, specic site malignancy (breast) has
increased association with glargine
(34), but in the circumstance of no prior
specic hypothesis and with multiple
sites t ested statistically without adjust-
ment, this too has very low evidence
impact.
A number of studi es have attempted
to address this area of investig ation, and
atte ntion has been drawn to their li mita-
tions (36 38). Boyle and colleagues
performed a detailed meta-analysis of
the insulin glargine observatio nal data.
This was presented at the International
Diabetes Federat ion annual meeting
(2011) and American Diabetes Associa-
tion Scientic Sessions (20 12), and the
presentations are available online
(39,40). As such, t hey are no t subject to
formal prior peer review, while the ac tiv-
ity was sponsored by the manufacturer of
insulin glargine. Nevertheless, the analy-
sis is comprehensive, and sensitivi ty anal-
yses address such issues as allowing the
effect of the Hemkens and colleagues dose
adjustment to be understood within the
context of the other published ndings.
Other studies presented in symposia at
the American Diabetes Association Scien-
tic Sessions 2012 from a U.S. insurance
company and the Kaiser Permanente da-
tabase reached similar conclusions: that
there is no associ ation with overall malig-
nancy or with any specictumorsite,
although even with meta-analysis there
are limitations in the amount of data avail-
able for breast or any other single site can-
cer, and of co urse du rati on of exposure is
limited by the introduction of insulin
glargine from 2003. How s uch limitations
should affect clinical pra ctice is discussed
below.
RCTs
Methodological issues
It might seem as though, as the gold
standard of treatment comparisons, RCTs
should be able to answer the questions
posed here. However, there are also issues
with the RCTs in this area. The most basic
of these is that all but one of the studies
(Outcome Redu ction With Initi al Glar-
gine Intervention [ORIGIN]) (see below)
were not set up to assess cancer outcomes,
which thus were for the most part col-
lected as serious adverse event (SAE)
repo rts. In areas such as cardiovas cular
disease, this is recognized as being prob-
lematic, as investigators m ay not be sure
what constitutes an event and may have
limited access to diagnostic information
from other medical servic es, perhaps
from health events that happened to the
pati ent in other cities. For malignancies,
this may n ot seem to be such a problem,
but w hen trawling through SAE and AE
data it becomes obvious that there are
problems with materials originating from
study sites
dnot least in the diverse ter-
minology used. Despite specication that
malignancies should be SAEs, some are
still reported as a dverse events (mi xed in
with a large number of benign tumors),
and the use of words like neoplasm of-
ten seems to be investigator solutions to
not knowing the nature of a lesion. Liver
metastases (or merely deposits on scans)
give similar problems as to origin. Only
rarely is some kind of post hoc adjud ica-
tion of m alignant events und ertaken, of-
ten without recourse to retrospective
inquiry for more details from investig ator
sites (a process t hat anyway is usually very
unrewarding). Additionally, many RCTs
are phase 3 studies of duration of ,12
months, which may not give time for
any drug effect to become manifested
and often contribut e less than three
events per study.
Meta-analysisisinsomeminds
given a magic al aura to deal with the
problems of RCTs. This is mostly not the
case, as meta-analysis is only as good as
the underlying data it considers, and
problems such as heterogeneity of the
S242 DIABETES CARE, VOLUME 36, SUPPLEMENT 2, AUGUST 2013 care.diabetesjournals.org
Insulin therapy and cancer
populations studied may even worsen
that compared with the better RCTs
alone. Ultimately, meta-analysis is only
useful for the purpose of gaining power
where the patient exposure i n available
studies is smalldoften thus b eing based
largely on the short-term phase 3 studies
with all their limitations.
RCT ndings
However, the ORIGIN study, an RCT of
insu lin glargine ver sus standa rd care in
persons ranging from prediabe tic to tak-
ing two oral agents at entry, is useful in
being of relatively high exposure, having
sizeable pa rticipant numbers followed
for a median of 6.2 years, and having
adjudicated cancer outcomes (41). By
studying insulin glargine, it effectively
deal s with the issues of both insulin and
that specic analog. The ndings were of
no benet or harm, with large numbers of
events (953) and thus tight CIs for all can-
cer (95% CI 0.881.13) and deaths from
malignancy (0.771.15) versus a mix of
other glucose-lowering therapies used
slightly less intensively. For specicma-
lignanci es, the results are inevitably less
certain owing to smaller numbers, but
the spread of ndings on either side of a
hazard ratio o f unity po ints to no identi-
able signal. Even for quite larg e CIs (say,
up to an 80% in creased risk as chosen by
the U.S. Food and Drug Administration
for cardiovascular events when licensing
new medications), statistical power re-
quires event numbers of .100 and ab-
sence of multiple testing, neither of
which can be expected to be att ained in
our current RCTs for any one organ sit e
malignancy. The weakness of the study is
that diabetes duration at entry was rela-
tively shor t (indeed, a small proportion of
particip ants had just glucose intolerance)
andBMInotashighasinsometreated
populations (mean ,30.0 kg/m
2
), so re-
sulting insulin dose in the treated group
was modest, even in those conti nuing in-
sulin treatment at 6 years (end of study
mean ;40 units/day). Accordingly, the
study does not deal with the question of
high exogenous insulin doses added to
high endogenous secretion in the more
markedly obese person.
A me ta-analysis of the insulin glargine
studies r epor ted ndings in both types of
diabetes, against NPH insulin, and in both
longer- and shorter-du ration studies (42) .
The ap proach has all of the problems dis-
cussed above, with only one study (the
reti nopathy study ) having duration of di-
abetes .1 year and that with limited
participant numbers (n = 514 exposed)
(43). Accor dingly, with only 91 persons
(glargin e and comparator) experiencing a
new malignancy, the upper CI was 1.36,
while for specic site malignanc ies such
as breast cancer the upper CI was .2.0.
There was no signal fo r an increase in ma-
lignancy in the retinopathy study. Boyle
and colle agues later extended these anal-
yses of the RCTs reporting hazard ratios of
0.88 (95% CI 0.800.97) for glargine versus
comparators, but suggested caution should
be exercised in overinterpreting the results
as indicating a small benet (39,40).
CONCLUSIONSdPresently, it is dif-
cult to conclude that there is any evi-
dence of risk of m alignancy from
exogenous insulin of a size great enough
to modify clinical practice decisions. Fur-
ther studies mi ght be directed at people
who have already had one neoplasm (and
therefore have a particularly hig h back-
ground ri sk), those with high background
risk for other reasons (e.g., genetic
makeup predisposing to breast cancer,
expo sure to asbestos), and those women
whose breast cancers overexpress insulin
receptor subtypes and IGF-1 receptors.
However, the issue of high-dose insulin
therapy has not been properly studied,
and such people nearly always have high
endo genous insulin secretio n and some
tendency to an enhanced inammatory
state, putting them at risk. It cannot
presentl y be argued that any of the groups
of people at higher risk should be denied
the benets of insulin therapy if they need
it, particularly as nearl y all other glucose-
lowering therapies have other putative or
real safety risks.
For insulin analogs, it mig ht s eem
unwise to introduce and develop another
diarginyl i nsuli n where other approaches
are possible, but insulin glarginetype
molecules seem safe . An issue will arise
if small-molecule insulin receptor ago-
nists are develo ped: these might end up
missignaling down the mitogen-activ ated
protein kinase pathway as insulin Asp-
B10 was presumed to do and, as such,
will need careful and fu ll preclinical and
toxicological review, including avoidance
of insulin receptor A binding. This will
not be an easy undertaking, as overdosing
of insulin kills animals from hypoglyce-
mia, limiting the use of conventional tox-
icologic al studies.
AcknowledgmentsdP.H. or institutions with
which he is connected receive funding from all
insulin manufacturers marketing products in
Europe and North America for his advisory,
education, and research activities. No other
potential conicts of in teres t releva nt to this
article were reported.
P.H. is resp onsible for the entire content of
the article, which was pre pared without ref-
erence to any other party excepting Diabetes
Care reviewers and editors.
References
1. Giovannucci E, Harlan DM, Archer MC,
et al. Diabetes and cancer: a consensus re-
port. Diabetes Care 2010;33:16741685
2. Yang X-L, Chan JCN. Diabetes, insulin
and cancer risk. World J Diabetes 2012;3:
6064
3. Larsson SC, Orsi ni N, Wolk A. Diabetes
mellitus and risk of color ectal c ancer:
a meta-analysis. J Natl Cancer Inst 20 05;
97:1 6791687
4. Oleksiewicz MB, Southgate J, Iversen L,
Egerod FL. Rat urinary bla dder carcino-
genesis by dual-acting PPARalpha + gamma
agonists. PPAR Res 2008;2008:103167
5. Dorman dy J, Bhattacharya M, van
Troostenburg de Bruyn A-R; PROactive
investigators. Saf ety and tolerability of
piogli tazone in high-risk patients with
type 2 diabe tes: an overview of data f rom
PROactive. Drug Saf 2009; 32:1 87202
6. Lewis JD, Ferrara A , Peng T, e t al. Risk of
bladder cancer among diabetic patients
treated with pioglitazone: interim report
of a longitudinal cohort study. Diabetes
Care 2011;34:916922
7. Djeli
c N. Analysis of sister-chromatid ex-
changes and micronuclei in cultured hu -
man lymphocytes treated with insulin.
Folia Biol (Praha) 2001;47:2831
8. Kurtzhals P, Schäffer L, Sørensen A, et al.
Corr elations of receptor binding and
metabolic and mitogenic potencies of in-
sulin analogs designed for clinical use.
Diabetes 2000;49:9991005
9. Frasc a F, Pandini G, Scali a P, et al. Insulin
receptor isoform A, a newly recognized,
high-afnity insulin-like growth factor II
receptor in fetal and cancer cells. Mol Cell
Biol 1999;19:32783288
10. Belore A, Frasca F. IGF and insulin receptor
signaling in breast cancer. J Mammary Gland
Biol Neoplasia 2008;13:381406
11. Sommerfeld MR, M
uller G, Tschank G, et al.
In vitro metabolic and mitogenic signaling
of insulin glargine and its metabolites. PLoS
ONE 2010;5:e9540
12. Mulligan AM, OMalley FP, Ennis M,
Fantus IG, Goodwin PJ. Insulin receptor is
an independent predictor of a favorable
outcome in early stage breast cancer. Breast
Cancer Res Treat 2007;106:3947
13. Loeper S, Ezzat S. Acromegaly: re-think-
ing the cancer risk. Rev Endocr Metab
Disor d 2008;9:4158
14. Papa V, Pezzino V, Costantino A, et al. El-
evated insulin receptor content in human
care. diabe tesjournals.org DIABETES CARE, VOLUME 36, SUPPLEMENT 2, AUGUST 2013 S243
Home
breast cancer. J Clin Invest 1990;86:1503
1510
15. Cullen KJ, Yee D, Sly WS, et al. Insulin-
like gro wth factor receptor expression and
function in human breast cancer. Cancer
Res 1990;50:4853
16. Drej er K, Kruse V, Larsen UD, Hougaard
P, Bjørn S, Gammeltoft S. Receptor bi nd-
ing and tyrosine kinase activation by in-
sulin analogues with extreme a fnities
studied in human hepatoma HepG2 cells.
Diabetes 1991;40 :14881495
17. Berti L, Kellerer M, Bossenmaier B, Seffer
E, Seipke G, Häring HU. The long acting
human insulin a nalog HOE 901: charac-
teristics of insulin signalling in com-
pari son to Asp(B10) and regular insulin.
Horm Metab Res 199 8;30:123129
18. Sommereld MR, M
uller G, Tschank G,
et al. In vitro metabolic and mitogenic sig-
nalling of insulin glargine and its metabolites
(Abstract). Diabetes 2010;59(Suppl. 1):A153
19. Bolli GB, Hahn AD, Schmidt R, et al. Plasma
exposure to insulin glargine and its me-
tabolites m1 and m2 after subcutaneous
injection of therapeutic and supratherapeutic
doses of glargine in subjects with type 1
diabetes. Diabetes Care 2012;35:2626
2630
20. Lucidi P, Porcellati F, Rossetti P, et al. Me-
tabolism of insulin glargine after repeated
daily subcutaneous injections in subjects
with type 2 diabetes. Diabetes Care 2012;
35:26472649
21. Bowker SL, Majumda r SR, Veugelers P,
Johnson JA. Increased cancer-related
mortality for patients with type 2 diabetes
who use sulfonylureas or insulin. Diabetes
Care 2006;29:254258
22. Yang X, Ko GTC, So WY, et al. Associa-
tions of hyperglycemia and insuli n usage
with the risk of cancer in type 2 diabetes:
the Hong Kong diabetes registry. Diabetes
2010;59:12541260
23. Johnson JA, Gale EAM. Diabetes, insulin
use, and cancer risk: are observational
studies part of the solution-or part of the
problem? Diabete s 2010;59:11291131
24. Johnson JA, Bowker SL, Richardson K, Marra
CA. Time-varying incidence of cancer after the
onset of type 2 diabetes: evidence of potential
detection bias. Diabetologia 2011;54:2263
2271
25. Currie CJ, Poole CD, Jenkins-Jones S,
Gale EAM, Jo hnson JA, Morgan CL.
Mortality after incident cance r in people
with and without type 2 diabetes: impact
of metformin on survival. Diabetes Care
2012;35:299304
26. Lind M, Fahlén M, Eliasson B, Odén A. The
relationship between the exposure time of
insulin glargine and risk of breast and
prostate cancer: an observational study of
the time-dependent effects of antidiabetic
treatments in patients with diabetes. Prim
Care Diabetes 2012;6:5359
27. Stevens RJ, Ali R, Bankhead CR, et al.
Canc er outcomes and a ll-cause mortalit y
in adults allocated to metformin: system-
atic review and collaborative meta-analysis
of randomised clinical trials. Diabetologia
2012;55:25932603
28. Yang XL, Ma RCW , So W-Y, Kong APS,
Xu G, Chan JCN. Addressing different
biases in analysing drug use on cancer risk
in diabete s in non-clinical trial settingsd
what, why and how? Diabetes Obes Metab
2012;14:579585
29. Suissa S, Azoulay L. Metformin and the
Risk of Cancer: Time-related biases in ob-
servational studies. Diabetes Care 2012;35:
26652673
30. Boyle P, Boniol M, Koechlin A, et al. Di-
abetes and breast cancer risk: a meta-
analysis. Br J Cancer 2012;107:16081617
31. Ghosal S, Stephens J, Deventer AV, et al.
Critical appraisal of the recent data published
on the link between insulin and cancer.
Diabetes Metab Res Rev 2011;5:211213
32. Hemkens LG, Grouven U, Bender R, et al.
Risk of malignancies in p atients with di-
abetes treated with human insulin or insulin
analogues: a cohort stu dy. Diabetologia
2009;52:17321744
33. Jonasson JM, Ljung R, Talbäck M,
Haglund B, Gudbjörnsdòttir S, S teineck
G. Insulin glargine use and short-term
incidence of malignancies-a population-
based f ollow-up study in Sweden. Dia-
betologia 2009;52:17451754
34. Colhoun HM; SDRN Epidemiology Group.
Use of insulin glargine and cancer incidence
in Scotland: a study from the Scottish Di-
abetes Research Network Epidemiology
Group. Diabetologia 2009;52:17551765
35. Currie CJ, Poole CD, Gale EA M. The in-
uenc e of glucose-lowering therapies on
cancer risk in type 2 diabetes. Dia-
betologia 2009;52:17661777
36. Suissa S, Azoulay L, Del lAniello S, Evans
M, Vora J, Pollak M. Long-term effects of
insulin glargine on the risk of breast can-
cer. Diabetologia 2011;54:22542262
37. Morden NE, Li u SK, Smith J, Mackenzie
TA, Skinner J, Korc M. Further explora-
tion of the relation ship between insulin
glargine and incident cancer: a retrospective
cohort study of older Medicare patients.
Diabetes Care 2011;34:19651971
38. Badrick EL, Buchan I, Renehan AG.
Comment on: Morden et al. Further explo-
ration of the relationship between insulin
glargine and incident cancer: a retrospective
cohort study of older Medicare patients.
Diabetes Care 2011;34:1965-1971. Diabetes
Care 2012;35:e15
39. Boyle P. Ins ulin glargine and cance r risk:
latest data [article online], 2011. Inter-
national Diabetes Federation Co ngress,
Dubai. A vailable from http://www.idf.org/
worlddiabetescongress/duba i-2011. Accessed
30 August 2012
40. Boyle P, Koechlin A, Boni ol M, Robertson
C, Bolli GB , Rosenstock J. Updated meta-
analysis of cancer risk among users of in-
sulin glargine (Abstract). Diabetes 2012;
61(Suppl. 1):A345
41. Gerst ein HC, Bosch J, Dagenais GR, et al.;
ORIGIN Trial Investigators. Basal insulin
and cardiovascular and other outcomes in
dysglycemia. N Engl J Med 2012;3 67:
319328
42. Home PD, Lagarenne P. Combined rando-
mised controlled trial experience of malig-
nancies in studies using insulin glargine.
Diabetologia 2009;52:24992506
43. Rosenstock J, Fonseca V, McGill JB, et al.
Simil ar risk of malign ancy with insuli n
glargine and neutral protamine Hagedorn
(NPH) in sulin in patients with type 2 di-
abetes: ndings from a 5 year randomised,
open-label study. Diabe tologia 20 09;52:
19711973
S244 DIABETES CARE, VOLUME 36, SUPPLEMENT 2, AUGUST 2013 care.diabetesjournals.org
Insulin therapy and cancer
... 21 A number of observational randomized trials were performed to determine if insulin therapy could be associated with increased risk of cancer in individuals with diabetes. [22][23][24] The outcome of the results were inconsistent, some reporting that insulin therapy is associated with increased risk of cancer while others showed no relevance to cancer risk. [22][23][24] The conflicting results of existing studies could reflect the biological diversity of cancer. ...
... [22][23][24] The outcome of the results were inconsistent, some reporting that insulin therapy is associated with increased risk of cancer while others showed no relevance to cancer risk. [22][23][24] The conflicting results of existing studies could reflect the biological diversity of cancer. In general, the data from epidemiological studies negate the effects of insulin therapy on cancer risk in individuals with type 2 diabetes. ...
Article
Full-text available
Background Non-melanoma skin cancer (NMSC) is a frequent type of malignancy with a steadily increasing incidence rate worldwide. Although NMSC was shown to be associated with diabetes, no studies have addressed the extent to which insulin use influences the risk of NMSC in light of social determinants of health (SDOH). We conducted a quantitative study that examined the interplay between insulin use, SDOH, additional covariates, and NMSC among individuals with diabetes. Methods We based our analysis on the 2020 Behavioral Risk Factor Surveillance System (BRFSS), a national survey conducted yearly in the US. We performed weighted chi-squared test, logistic regression, and survival analyses on 8685 eligible participants with diabetes enrolled in the BRFSS. Results Kaplan Meier survival curves showed higher probability of NMSC event-free survival for participants with diabetes using insulin compared to participants with diabetes not using insulin (log-rank test P < .001). Significant associations were detected between insulin use and reduced odds of NMSC (OR .56; 95% CI: .38-.82), and decreased hazard (HR .36; 95% CI: .21-.62), along with indices of SDOH. Conclusions Our findings suggest that socioeconomic differences related to the healthcare system and behavioral patterns are linked to discrepancies in the use of insulin and the development of NMSC.
... More specifically, insulin has been shown to activate members of the insulin-like growth factor (IGF) receptor family to inhibit apoptosis and subsequently prolong the survival of these transformed breast tissue cells (125). This mechanism suggests that insulin not only facilitates glucose uptake but may also support a tumour-friendly microenvironment, thereby affecting whole-body glucose metabolism and contributing to cancer progression (126). This interplay between insulin, glucose metabolism, and cancer risk underscores the importance of monitoring insulin therapy and metabolic health in cancer-prone populations. ...
Article
Full-text available
Recent discoveries demonstrated the skeleton’s role as an endocrine organ regulating whole-body glucose homeostasis. Glucose metabolism is critical for rapid cell proliferation and tumour growth through increasing glucose uptake and fermentation of glucose to lactate despite being in an aerobic environment. This hypothesis paper discusses emerging evidence on how bones can regulate whole-body glucose homeostasis with potential to impact on tumour growth and proliferation. Moreover, it proposes a clinical link between bone glucose metabolism and prognosis of cancer based on recent clinical trial data. Targeting metabolic pathways related with classic glucose metabolism and also bone metabolism, novel methods of cancer therapy and treatment could be developed. This paper objective is to highlight the need for future research on this altered metabolism with potential to change future management of cancer patients.
... Insulin is a growth factor, and high levels of endogenous insulin could stimulate neoplastic growth. 27,28 Notably, a systematic review/metaanalysis reported that exogenous insulin was associated with increased pancreatic cancer risk in patients with diabetes mellitus. [29][30][31][32][33] Insulin promotes pancreatic cancer initiation and sustains pancreatic cancer development by eliciting tumor proliferation through the activation of Akt/PI3K pathway. ...
Article
Full-text available
Extrahepatic malignancies are the leading cause of death in patients with nonalcoholic fatty liver disease (NAFLD). Of these cancers, pancreatic cancer is one of the most lethal; however, the link between NAFLD and pancreatic cancer remains unclear. Recently, various research results have been reported on the association between NAFLD and pancreatic cancer, and the results of compiling this information revealed the following. First, the prevalence of pancreatic cancer in patients with NAFLD is at 0.26%. Second, the currently evident pathogenesis includes intrapancreatic risk factors, such as: (1) non‐alcoholic fatty pancreas disease, and (2) intraductal papillary mucinous neoplasm; and extrapancreatic risk factors, such as: (1) insulin resistance and adipocytokines, (2) proinflammatory cytokines, and (3) dysbiosis. Finally, metformin and sodium–glucose cotransporter 2 inhibitors may reduce the risk of pancreatic cancer in diabetes patients with NAFLD. In this review, we summarize the recent evidence on the epidemiology and mechanisms for NAFLD‐related pancreatic cancer. We further discuss the impact of anti‐diabetic medication on pancreatic cancer.
... Additionally, it aids in storing excess glucose in the liver for future use, contributing to maintaining blood sugar levels within a healthy range. [96], [97], [98], [99], [100] Pancreatic enzymes constitute a cluster of digestive enzymes crucial for breaking down food in the digestive system. While not commonly employed as a direct treatment for pancreatic cancer, they find utility in managing symptoms associated with pancreatic insufficiency resulting from pancreatic diseases or treatments. ...
Preprint
Full-text available
Background: Malignant neoplasm of the pancreas (MNP), a highly lethal illness with bleak outlook and few therapeutic avenues, entails numerous cellular transformations. These include irregular proliferation of ductal cells, activation of stellate cells, initiation of epithelial-to-mesenchymal transition, and changes in cell shape, movement, and attachment. Discovering potent drug cocktails capable of addressing the genetic and protein factors underlying pancreatic cancer's development is formidable due to the disease's intricate and varied nature. Method: In this study, we introduce a fresh model utilizing Graph Attention Networks (GATs) to pinpoint potential drug pairings with synergistic effects for MNP, following the RAIN protocol. This protocol comprises three primary stages: Initially, employing Graph Neural Network (GNN) to suggest drug combinations for disease management by acquiring embedding vectors of drugs and proteins from a diverse knowledge graph encompassing various biomedical data types, such as drug-protein interactions, gene expression, and drug-target interactions. Subsequently, leveraging natural language processing to gather pertinent articles from clinical trials incorporating the previously recommended drugs. Finally, conducting network meta-analysis to assess the relative effectiveness of these drug combinations. Result: We implemented our approach on a network dataset featuring drugs and genes as nodes, connected by edges representing their respective p-values. Our GAT model identified Gemcitabine, Pancrelipase Amylase, and Octreotide as the optimal drug combination for targeting the human genes/proteins associated with this cancer. Subsequent scrutiny of clinical trials and literature confirmed the validity of our findings. Additionally, network meta-analysis confirmed the efficacy of these medications concerning the pertinent genes. Conclusion: By employing GAT within the RAIN protocol, our approach represents a novel and efficient method for recommending prominent drug combinations to target proteins/genes associated with pancreatic cancer. This technique has the potential to aid healthcare professionals and researchers in identifying optimal treatments for patients while also unveiling underlying disease mechanisms.
... Insulin, a hormonal molecule, can lower blood sugar and acts as a potent factor that in uences cell proliferation [31]. Insulin and its precursors are powerful cell proliferation promoters. ...
Preprint
Full-text available
Background Current epidemiological data show that diabetes, especially type 2 diabetes, is considered to be a complicated factor in the prevalence and prognosis of prostate cancer. Methods The computer retrieves four databases to obtain controlled trials at home and abroad on the efficacy and prognostic effect of metformin in patients with diabetes and prostate cancer. After a rigorous literature quality evaluation, we utilized the RevMan 5.3 software for analyzing the data. Results We included the 8 studies for this comprehensive meta-analysis. These studies were considered to be controlled or array trials. For comparing the test group and the control group, we collected the data recurrence rate from the 5 studies. We revealed that the 5-year recurrence rate was not statistically different (OR: 0.92; 95% Cl: 0.67,1.28; P = 0.631). Similarly, we found the 10-year recurrence rate (OR:0.81; 95% Cl: 0.25,2.58; P = 0.715), 5-year mortality rate (OR:0.66; 95% Cl: 0.14,3.11; P = 0.597), and 10-year mortality rate (OR: 0.44; 95% Cl: 0.26,0.74; P < 0.01). Although we found the asymmetrically distributed effective rate in the funnel plot analysis, publish bias was not potentially found in Egger’s test (P = 0.958). Conclusion This meta-analysis demonstrated that metformin is not potentially effective in patients with diabetes and prostate cancer, as evidenced by the 5-year recurrence rate, 10-year recurrence rate, 5-year mortality, and 10-year mortality, and the above conclusions need to be verified by more high-quality research.
... Patients with diabetes who are on insulin therapy are typically exposed to high levels of exogenously administered insulin to achieve optimal glucose control [17]. Therefore, it is biologically plausible that treatment with insulin increases risk of cancer since insulin is a growth factor that can stimulate neoplastic growth [96,97]. Earlier observational studies reported concerning results about the plausible link between insulin use and risk of cancer; however, many of these studies failed to take into account dose, duration, and timing of insulin exposure, so it was not possible to draw firm conclusions [98][99][100][101]. ...
Article
Full-text available
Purpose of Review Diabetes is associated with an increased risk for several types of cancer. Therefore, use of antihyperglycemic medications to lower blood glucose may modify cancer risk. Here we review available data on the link between the most common classes of antihyperglycemic agents and cancer risk among patients with diabetes. Recent Findings A database search was conducted between February 2022 and June 2022 on PubMed and Embase for systematic reviews and meta-analyses investigating the association between antihyperglycemic agents and risk of cancer. Use of biguanides such as metformin is associated with 20–30% lower risk for all cancer incidence, and somewhat greater benefit for cancer-related mortality. Alpha-glucosidase inhibitors, e.g., acarbose, have not been consistently associated with cancer. Similarly, no consistent effects have been reported for thiazolidinediones, but the relationship with cancer seems to depend on the type of drug, dose, and duration of treatment. Exposure to various types of incretin-based therapies (glucagon-like peptide-1 agonists and dipeptidyl peptidase-4 inhibitors) has not been found to significantly modify cancer risk. Inhibitors of sodium glucose cotransporter-2 may raise risk for bladder cancer and reduce risk for gastrointestinal cancer. Use of insulin and insulin analogs is associated with a significant increase in total cancer risk by almost 50% compared to other antihyperglycemic drugs. Likewise, insulin secretagogues like sulfonylureas have generally been linked to greater risk for cancer by ~ 20%, although these associations may be agent-specific and dose-dependent. Summary Current evidence suggests that the risk of cancer associated with the use of antihyperglycemic medications among patients with diabetes depends on the class of drug and type of agent, dosage, and duration of treatment. More research is needed to delineate the mechanisms by which these agents affect the process of carcinogenesis.
... Different antidiabetic drugs have different effects on risk of cancer. Metformin has been reported to decrease cancer risk or cancer mortality [13], whereas insulin and sulfonylureas might be associated with increased cancer risk [14,15]. The results for thiazolidinediones are controversial, which may increase [16], decrease [17], or have a neutral effect [18] on the risk of cancer or cancer progression. ...
Article
Full-text available
Diabetes and cancer are common diseases and are frequently diagnosed in the same individual. These patients need to take antidiabetic drugs while receiving antitumor drugs therapy. Recently, immunotherapy offers significant advances for cancer treatment. However, it is unclear whether antidiabetic drugs affect immunotherapy. Here, by employing syngeneic mouse colon cancer model and melanoma model, we studied the effects of 6 common antidiabetic drugs on anti-PD1 immune checkpoint inhibitor in tumor treatment, including acarbose, sitagliptin, metformin, glimepiride, pioglitazone, and insulin. We found that acarbose and sitagliptin enhanced the tumor inhibition of anti-PD1, and metformin had no effect on the tumor inhibition of anti-PD1, whereas glimepiride, pioglitazone, and insulin weakened the tumor inhibition of anti-PD1. Our study suggests that cancer patients receiving anti-PD1 antibody therapy need serious consideration when choosing antidiabetic drugs. In particular, acarbose significantly inhibited tumor growth and further enhanced the therapeutic effect of anti-PD1, which can be widely used in tumor therapy. Based on this study, further clinical trials are expected.
... Controlling these prevents and/or delays the development of cardiovascular disease [11][12][13]. 71% are overweight or obese, a chronic disease, which is associated with other cardiovascular risk factors and cancer [14][15][16][17][18][19]. Despite the existence of the obesity law, the coverage of its treatment has many drawbacks. ...
Article
Full-text available
Diabetes mellitus (DM) is a significant risk factor for various cancers, with the impact of anti-diabetic therapies on cancer progression differing across malignancies. Among these therapies, metformin has gained attention for its potential anti-cancer effects, primarily through modulation of the AMP-activated protein kinase/mammalian target of rapamycin (AMPK/mTOR) pathway and the induction of autophagy. Beyond metformin, other conventional anti-diabetic treatments, such as insulin, sulfonylureas (SUs), pioglitazone, and dipeptidyl peptidase-4 (DPP-4) inhibitors, have also been examined for their roles in cancer biology, though findings are often inconclusive. More recently, novel medications, like glucagon-like peptide-1 (GLP-1) receptor agonists, dual GLP-1/glucose-dependent insulinotropic polypeptide (GIP) agonists, and sodium-glucose co-transporter-2 (SGLT-2) inhibitors, have revolutionized DM management by not only improving glycemic control but also delivering substantial cardiovascular and renal benefits. Given their diverse metabolic effects, including anti-obesogenic properties, these novel agents are now under meticulous investigation for their potential influence on tumorigenesis and cancer advancement. This review aims to offer a comprehensive exploration of the evolving landscape of glucose-lowering treatments and their implications in cancer biology. It critically evaluates experimental evidence surrounding the molecular mechanisms by which these medications may modulate oncogenic signaling pathways and reshape the tumor microenvironment (TME). Furthermore, it assesses translational research and clinical trials to gauge the practical relevance of these findings in real-world settings. Finally, it explores the potential of anti-diabetic medications as adjuncts in cancer treatment, particularly in enhancing the efficacy of chemotherapy, minimizing toxicity, and addressing resistance within the framework of immunotherapy.
Article
Type 1 diabetes mellitus (T1D) is a chronic disease with potentially severe complications, and β-cell deficiency underlies this disease. Despite active research, no therapy to date has been able to induce β-cell regeneration in humans. Here, we discover the β-cell regenerative effects of glucagon receptor antibody (anti-GcgR). Treatment with anti-GcgR in mouse models of β-cell deficiency leads to reversal of hyperglycemia, increase in plasma insulin levels, and restoration of β-cell mass. We demonstrate that both β-cell proliferation and α- to β-cell transdifferentiation contribute to anti-GcgR-induced β-cell regeneration. Interestingly, anti-GcgR-induced α-cell hyperplasia can be uncoupled from β-cell regeneration after antibody clearance from the body. Importantly, we are able to show that anti-GcgR-induced β-cell regeneration is also observed in non-human primates. Furthermore, anti-GcgR and anti-CD3 combination therapy reverses diabetes and increases β-cell mass in a mouse model of autoimmune diabetes.
Article
Full-text available
We appreciate the discussion initiated by Badrick et al. (1) and thank them for the questions raised. In an ideal world, we would have a randomized trial with the enormous sample necessary to answer this question as well as decades of follow-up. In an imperfect world, our analysis reveals the experience of a significant fraction of U.S. patients with diabetes using insulin glargine over 3 years of observation. Regarding our prevalent users cohort and fixed treatment group assignment, we acknowledged the reasonable but unproven assumption that our observed exposure reflects unobserved exposure, preceding 2006 (Part D inception). Under this assumption, assignment to treatment observed in the earliest observation window is the best exposure …
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
Objective: Time-related biases in observational studies of drug effects have been described extensively in different therapeutic areas but less so in diabetes. Immortal time bias, time-window bias, and time-lag bias all tend to greatly exaggerate the benefits observed with a drug. Research design and methods: These time-related biases are described and shown to be prominent in observational studies that have associated metformin with impressive reductions in the incidence of and mortality from cancer. As a consequence, metformin received much attention as a potential anticancer agent; these observational studies sparked the conduction of randomized, controlled trials of metformin as cancer treatment. However, the spectacular effects reported in these studies are compatible with time-related biases. Results: We found that 13 observational studies suffered from immortal time bias; 9 studies had not considered time-window bias, whereas other studies did not consider inherent time-lagging issues when comparing the first-line treatment metformin with second- or third-line treatments. These studies, subject to time-related biases that are avoidable with proper study design and data analysis, led to illusory extraordinarily significant effects, with reductions in cancer risk with metformin ranging from 20 to 94%. Three studies that avoided these biases reported no effect of metformin use on cancer incidence. Conclusions: Although observational studies are important to better understand the effects of drugs, their proper design and analysis is essential to avoid major time-related biases. With respect to metformin, the scientific evidence of its potential beneficial effects on cancer would need to be reassessed critically before embarking on further long and expensive trials.
Article
Full-text available
OBJECTIVE In vivo, after subcutaneous injection, insulin glargine (21(A)-Gly-31(B)-Arg-32(B)-Arg-human insulin) is enzymatically processed into 21(A)-Gly-human insulin (metabolite 1 [M1]). 21(A)-Gly-des-30(B)-Thr-human insulin (metabolite 2 [M2]) is also found. In vitro, glargine exhibits slightly higher affinity, whereas M1 and M2 exhibit lower affinity for IGF-1 receptor, as well as mitogenic properties, versus human insulin. The aim of the study was to quantitate plasma concentrations of glargine, M1, and M2 after subcutaneous injection of glargine in male type 1 diabetic subjects.RESEARCH DESIGN AND METHODS Glargine, M1, and M2 were determined in blood samples obtained from 12, 11, and 11 type 1 diabetic subjects who received single subcutaneous doses of 0.3, 0.6, or 1.2 U · kg(-1) glargine in a euglycemic clamp study. Glargine, M1, and M2 were extracted using immunoaffinity columns and quantified by a specific liquid chromatography-tandem mass spectrometry assay. Lower limit of quantification was 0.2 ng · mL(-1) (33 pmol · L(-1)) per analyte.RESULTSPlasma M1 concentration increased with increasing dose; geometric mean (percent coefficient of variation) M1-area under the curve between time of dosing and 30 h after dosing (AUC(0-30h)) was 1,261 (66), 2,867 (35), and 4,693 (22) pmol · h · L(-1) at doses of 0.3, 0.6, and 1.2 U · kg(-1), respectively, and correlated with metabolic effect assessed as pharmacodynamics-AUC(0-30h) of the glucose infusion rate following glargine administration (r = 0.74; P < 0.01). Glargine and M2 were detectable in only one-third of subjects and at a few time points.CONCLUSIONS After subcutaneous injection of glargine in male subjects with type 1 diabetes, exposure to glargine is marginal, if any, even at supratherapeutic doses. Glargine is rapidly and nearly completely processed to M1 (21(A)-Gly-human insulin), which mediates the metabolic effect of injected glargine.
Article
OBJECTIVE—Some preclinical in vivo studies and limited human data suggest a possible increased risk of bladder cancer with pioglitazone therapy. This is an interim report of an ongoing cohort study examining the association between pioglitazone therapy and the risk of bladder cancer in patients with diabetes. RESEARCH DESIGN AND METHODS—This study includes 193,099 patients in the Kaiser Permanente Northern California diabetes registry who were $40 years of age between 1997 and 2002. Those with prior bladder cancer were excluded. Ever use of each diabetes medication (defined as two or more prescriptions within 6 months) was treated as a timedependent variable. Cox regression–generated hazard ratios (HRs) compared pioglitazone use with nonpioglitazone use adjusted for age, sex, race/ethnicity, diabetes medications, A1C, heart failure, household income, renal function, other bladder conditions, and smoking. RESULTS—The group treated with pioglitazone comprised 30,173 patients. There were 90 cases of bladder cancer among pioglitazone users and 791 cases of bladder cancer among nonpioglitazone users. Overall, ever use of pioglitazone was not associated with risk of bladder cancer (HR 1.2 [95 % CI 0.9–1.5]), with similar results in men and women (test for interaction
Article
People with type 2 diabetes mellitus have an excess risk of macrovascular disease and a poorer prognosis. PROactive (PROspective pioglitAzone Clinical Trial In macro Vascular Events) was a landmark study of secondary cardiovascular disease (CVD) prevention in type 2 diabetes that suggested a beneficial effect of pioglitazone therapy on macrovascular outcomes. Previous studies have already shown that pioglitazone has a good safety and tolerability profile in people with type 2 diabetes, but PROactive provided an opportunity to assess tolerability and safety associated with long-term exposure in a vulnerable subpopulation at very high cardiovascular risk. This review discusses all the key safety and tolerability characteristics associated with pioglitazone therapy in PROactive. As in previous studies, pioglitazone was associated with typical, but manageable, increases in oedema (26.4% vs 15.1% for placebo) and weight gain (mean change of +3.8 kg vs −0.6 kg for placebo). Increased hypoglycaemia with pioglitazone was consistent with improved glycaemic control. Despite more reports of serious heart failure in the pioglitazone group (5.7% vs 4.1% for placebo), there was a proportional improvement in macrovascular outcomes among patients developing heart failure, and absolute rates of macro-vascular events and mortality were similar to those in the placebo group. Liver function tests confirmed the hepatic safety of pioglitazone with long-term use and revealed a tendency to improved hepatic function, which may reflect reductions in liver fat. The comparative incidence of malignancies was similar; however, more cases of bladder neoplasm (14 vs 5) and fewer cases of breast cancer (3 vs 11) were observed in the pioglitazone versus placebo arms of the study. A higher rate of bone fractures observed among pioglitazone-treated female patients (5.1% vs 2.5%) warrants further investigation. Overall, safety and tolerability was predictable, and adverse events were not treatment limiting. These results suggest that any beneficial effects of pioglitazone on macrovascular outcomes are accompanied by good long-term tolerability in this population of very high-risk patients with type 2 diabetes and established CVD.
Article
Recent publications indicate a possible association between newer analogue insulin Glargine and cancer. However all these data were observational in nature and subject to lot of methodological errors. It is practically impossible to derive at a cause and effect relationship based on these observational studies. There are several confounding factors like hyperinsulinemia, obesity and smoking, which independently can be associated with cancer risk. This review looks into the recently published observational studies from the methodological point of view and whether such a negative association can indeed be drawn as a conclusion. Several methodological defects were identified in the process of scrutiny and we concluded it is not possible to conclude that there is an association between Glargine use and cancer. However long-term prospective data is required to come to definite conclusion.