You are on page 1of 9

REVIEW ARTICLE

Mechanisms of Drug Resistance Related to the


Microenvironment of Solid Tumors and Possible
Strategies to Inhibit Them
Qian Tan, MD, MSc,* Jasdeep K. Saggar, MSc, PhD,* Man Yu, MSc, PhD,* Marina Wang, BSc,*
and Ian F. Tannock, MD, PhD*†‡
acids, leading to an acidic microenvironment in tumors (Fig. 1).
Abstract: Drug resistance can occur at the individual cellular level or as a The abnormal vascular structure and function of solid tumors also
result of properties of the tumor microenvironment. The convoluted vascu- limit the delivery of anticancer drugs.2–4
lature within tumors results in robustly proliferating well-nourished cells For a systemically administered anticancer drug to kill a high
located proximal to functional blood vessels and regions of slowly prolifer- proportion of cancer cells in a solid tumor, it must cross blood ves-
ating (often hypoxic) cells located distal to functional blood vessels. Irreg- sel walls and penetrate into tumor tissue. However, the distribution
ular blood flow and large distances between functional blood vessels in of many drugs within tumors is heterogeneous, so that only a pro-
solid tumors lead to poor drug distribution within them such that cells distal portion of the target tumor cells are exposed to a potentially lethal
from functional blood vessels are exposed to ineffective concentrations of concentration of the cytotoxic agent: regions distal to blood ves-
drug, resulting in therapeutic resistance. Strategies to improve or comple- sels receive lower amounts of drug than do those cells located
ment the distribution of anticancer drugs within tumors hold promise for proximal to tumor blood vessels. Within nutrient-deprived tumor
increasing antitumor effects without corresponding increases in normal tis- regions, the rate of tumor cell proliferation is relatively low,5,6
sue toxicity. In particular, use of hypoxia-targeted agents and modulation of and slowly proliferating cells are resistant to most currently used
autophagy have shown promising results in enhancing the distribution of anticancer drugs, including many targeted agents. Both of these
drug activity within solid tumors and hence antitumor efficacy. In this re- effects contribute to the relative resistance to anticancer drugs of
view, we describe causes of resistance to chemotherapy that relate to the cells located distal to functional blood vessels.
microenvironment of solid tumors and the potential to improve antitumor
effects by countering such mechanisms of resistance.
Tumor Hypoxia
Key Words: Autophagy, chemotherapy, drug distribution, drug resistance,
hypoxia The convoluted vasculature within tumors results in regions
where the oxygen concentration falls to zero.7 The distance from
(Cancer J 2015;21: 254–262) functional blood vessels to hypoxic regions will depend on the
rate of oxygen consumption by the tumor cells but is typically at
TUMOR MICROENVIRONMENT AND a distance greater than 70 μm.8 Cells in chronically hypoxic re-
DRUG RESISTANCE gions may be viable, but they are often adjacent to region of necro-
sis and are probably destined to die in the absence of treatment.
Acute hypoxia is also common in tumors and results from inter-
Tumor Vasculature mittent blood flow through tumor vessels.9,10
The tumor vasculature is a critical regulator of the delivery of Tumor hypoxia is an important mediator of resistance to can-
nutrients to tumor cells and of the removal of products of metab- cer treatment. Cellular sensitivity to radiation depends on the gen-
olism and consequently of tumor growth. Although tumor cells re- eration of free radicals in the presence of oxygen, so that hypoxic
lease factors that stimulate angiogenesis, tumor vasculature often cells are relatively radioresistant. Hypoxic cells may also be resis-
lacks an organized structure. As cancer cells proliferate, blood tant to many anticancer drugs because (i) drugs are unlikely to be
vessels may become separated by longer distances than in normal delivered in toxic concentrations to hypoxic regions; (ii) low con-
tissues, and raised interstitial fluid pressure (IFP) and a dense ex- centrations of nutrient metabolites in hypoxic regions inhibit can-
tracellular matrix may compress blood vessels and contribute to ir- cer cell proliferation, and slowly proliferating cells are resistant to
regular blood flow.1,2 Consequently, there is a limited distribution cycle-active drugs2; (iii) tumor hypoxia is linked to loss of the p53
of nutrients, including oxygen to cells located distally from func- tumor suppressor protein that may result in loss of apoptotic abil-
tional blood vessels. There may also be a buildup of cellular ity and to increased angiogenesis and invasiveness11,12; (iv) hyp-
breakdown products in such regions including lactic and carbonic oxic cells can down-regulate expression of DNA topoisomerase
II, so that drugs such as doxorubicin and etoposide that target this
protein will be inefficient13; (v) hypoxia up-regulates genes in-
From the *Department of Medical Biophysics, University Health Network,
University of Toronto; †Division of Medical Oncology and Hematology, Prin- volved in drug resistance, including MDR genes encoding the
cess Margaret Cancer Centre; and ‡University Health Network, University of drug export pump P-glycoprotein14; and (vi) hypoxia can induce
Toronto, Toronto, Ontario, Canada. autophagy, which is a survival mechanism of stressed cells.15,16
Conflicts of Interest and Source of Funding: Supported by grant KG100252
from the Komen Foundation and by grants from the Canadian Institutes of
Health Research. The authors have disclosed that they have no significant Tumor Acidity
relationships with, or financial interest in, any commercial companies
pertaining to this article. The extracellular pH in solid tumors is often lower than in nor-
Reprints: Ian F. Tannock, MD, PhD, and Qian Tan, MD, MSc, Princess Margaret mal tissues, because of a high rate of glycolysis with production of
Cancer Centre, 9th Floor Room 417, 610 University Ave, Toronto, Ontario, lactic and carbonic acids and poor clearance of these acids.17,18
Canada M5G 2M9. E-mail: ian.tannock@uhn.ca;
susie.tan@uhnres.utoronto.ca.
Hypoxia also up-regulates the expression and activity of carbonic
Copyright © 2015 Wolters Kluwer Health, Inc. All rights reserved. anhydrase, which leads to enhanced extracellular acidification.19
ISSN: 1528-9117 The acidic extracellular environment causes basic anticancer drugs

254 www.journalppo.com The Cancer Journal • Volume 21, Number 4, July/August 2015

Copyright © 2015 Wolters Kluwer Health, Inc. All rights reserved.


The Cancer Journal • Volume 21, Number 4, July/August 2015 Drug Resistance Related to Solid Tumors

FIGURE 1. The tumor microenvironment. Schematic representation of a functional blood vessel and its surrounding cells. Cells that are closest
to the blood vessel are richly nourished with oxygen and other nutrients and therefore proliferate rapidly. Cells that are farther away are
nutrient deprived and may become hypoxic and have an acidic microenvironment; they tend to proliferate slowly.

(such as doxorubicin) to be charged, resulting in poorer uptake into become less effective because less drug is available to enter the
cells because it is the uncharged form that diffuses across the nucleus to exert cytotoxic effects18; sequestration of drugs in these
cell membrane. compartments will also lead to less drug being available to diffuse
The strong pH gradient between the cytoplasm of tumor cells to cells more distant from functional blood vessels (Fig. 2). As
and acidic intracellular organelles such as endosomes and lyso- well as being sites of sequestration of basic drugs, acidic
somes20,21 may also be involved in resistance to anticancer drugs. endosomes are central to the process of autophagy, discussed be-
Many anticancer agents are weak lipophilic bases (e.g., doxorubi- low as a cause of drug resistance.
cin, mitoxantrone), and they will accumulate in acidic organelles. We have shown that agents that raise endosomal pH might
When drugs are sequestered in the organelles of tumor cells, they decrease endosomal sequestration of basic anticancer drugs and

FIGURE 2. Effects of endosomal sequestration of basic drugs (nucleus [green], endosome [red/gray]). Top panel, basic anticancer drugs are
concentrated and sequestered in acidic endosomes of cells; this can limit their toxicity because less drug goes into the nucleus. Lower panel,
Agents such as chloroquine or a PPI (e.g., pantoprazole) can increase endosomal pH, thereby decreasing endosomal sequestration of basic
anticancer drugs and allowing more drugs to enter the nucleus and exert their activity, and more drugs to diffuse to distal cells.

© 2015 Wolters Kluwer Health, Inc. All rights reserved. www.journalppo.com 255

Copyright © 2015 Wolters Kluwer Health, Inc. All rights reserved.


Tan et al The Cancer Journal • Volume 21, Number 4, July/August 2015

thereby modify their toxicity and tissue penetration.22 Increasing so that increase in membrane-bound LC3B-II can be due to either
endosomal pH might be achieved by concomitant use of other ba- increased LC3-I processing from activation of autophagy or a
sic compounds (such as chloroquine) or by using inhibitors of the buildup of membrane-bound LC3B-II following inhibition of ly-
proton pump that maintain endosomal pH at a low value (Fig. 2). sosomal fusion. Use of antibodies to determine activation or inhi-
This pump is related to that which maintains acidity in the stomach bition of autophagy using LC3B isoforms is difficult if only
and may be inhibited by proton pump inhibitors (PPIs), which are LC3B-II is used. An additional autophagy marker, p62/SQSTM1
used clinically to reduce gastric acidity. We and others have ob- (p62), is recruited with LC3B-II to autophagosomes but, unlike
tained evidence that PPIs may enhance the effects of chemotherapy LC3B-II, is degraded within the mature autolysosome.34,35 Thus,
in animal models.23–27 observation of increased p62 is indicative of a buildup of the protein
because of inhibition of lysosomal fusion to the autophagosome,
Autophagy that is, to inhibition of autophagy.
Autophagy is prognostic of poor outcome in multiple tu-
Autophagy is a cellular mechanism used to digest old or
mor types, including cancers of the breast, lung, colon, and
damaged cellular components into component residues, which
melanoma.36–39 High levels of autophagy have been associated
may be recycled to generate essential macromolecules. All cells
with resistance to systemic therapy in several preclinical and
undergo autophagy, but it is up-regulated in stressed cells such
clinical models, presumably because autophagy facilitates sur-
as those with nutrient or growth factor depletion and hypoxia;
vival of stressed or damaged cells through recycling of cellular
such stress is common in nutrient-deprived regions of solid tu-
breakdown products.40 We have shown that treatment of cancer
mors, and autophagy colocalizes with hypoxia in tumors.16,28 Au-
cells in vitro and of solid tumors in mice with a wide variety of
tophagy involves the formation of autophagosomes, which have a
anticancer agents induces autophagy, suggesting that it is a
double membrane enclosing cytoplasmic cellular components;
common survival mechanism for drug-damaged cells, espe-
these then fuse with lysosomes to produce mature autolysosomes
cially those where autophagy is already up-regulated because
in which cellular proteins are degraded by cathepsins29–31 (Fig. 3).
of nutrient deprivation and therefore an important cause of
A series of autophagy-related proteins (known as ATGs) are
drug resistance.
responsible for the induction and regulation of autophagy,32 and
some of them are used as markers of autophagy that can be quan-
tified in Western blots or by immunohistochemistry (IHC) applied Drug Distribution in Solid Tumors
to tumor sections. The human form of ATG8 is microtubule- Drugs exit blood vessels and penetrate into tissues by con-
associated protein light-chain 3B (LC3B), which exists in a cyto- vection and/or diffusion. Convection depends on pressure gradi-
solic form as LC3B-I. Upon activation of autophagy, LC3B-I is ents between the vascular space and the interstitial space, vessel
cleaved and modified to LC3B-II, which then binds to the mem- permeability and the surface area for exchange, and the volume
brane of the autophagosome. Because of its direct association and structure of the extracellular matrix; it is most important for
with the autophagosome, LC3B-II has been used widely as a transport of large molecules.41 Drug diffusion is determined by
marker of autophagy in mammalian models.33,34 However, concentration gradients and by the size and charge of the mole-
LC3B-II can undergo turnover after fusion of the lysosome,33,34 cules and is the dominant mechanism for drugs of smaller

FIGURE 3. Schematic diagram of autophagy in a eukaryotic cell including the role of p62 and LC3B. Autophagy is induced by deprivation of
nutrients, hypoxia, and stress to form a membrane-contained phagophore. The second step entails sequestering of cellular breakdown
products in the double-membrane autophagosome. During this step, the LC3B protein is recruited to the auophagosome and p62 along with
the attached ubiquitin cargo attached to the LC3B protein. The fusion of a lysosome with an autophagosome generates the autolysosome.
At this step, LC3B disassociates, and p62 is degraded.

256 www.journalppo.com © 2015 Wolters Kluwer Health, Inc. All rights reserved.

Copyright © 2015 Wolters Kluwer Health, Inc. All rights reserved.


The Cancer Journal • Volume 21, Number 4, July/August 2015 Drug Resistance Related to Solid Tumors

molecular weight.42 Diffusion of drugs is also limited by binding have shown steep gradients of decreasing drug concentration
in tissue or by their rapid metabolism once they have extravasated. with increasing distance from tumor blood vessels and low
An important determinant of drug distribution within tissues is the penetration of drugs into hypoxic areas.56,57 For example,
half-life of the drug in the circulation; drugs that have a long half- doxorubicin intensity decreases to half at approximately 40 to
life have a better opportunity to achieve equilibrium within the 50 μm from the nearest blood vessels such that many viable cells
tumor microenvironment.43 are not exposed to detectable concentrations of drug after a single
Drug distribution can be evaluated in cell culture systems injection (Fig. 5A).
and in experimental solid tumors. In vitro multicellular mo- Most anticancer drugs are nonfluorescent, so their distribu-
dels include tumor spheroids and multilayered cell cultures tion within tumor tissues is difficult to assess. Fluorescently
(MCCs).44–47 Spheroids develop hypoxic areas as well as cen- tagged antibodies can recognize some drugs, such as the mono-
tral areas of necrosis once they grow to ~500 μm in diameter.48 clonal antibodies cetuximab and trastuzumab; these agents have
Drug distribution in spheroids can be studied for fluorescent an initial poor distribution, but equilibrium is then reached
drugs or by using autoradiography to determine the distribution throughout the tumor, most likely because of their long half-
of labeled drugs.49,50 Experiments using spheroids have shown lives in the circulation.58 The distribution of activity of other drugs
steep gradients of drug concentration from the surface for doxo- can be evaluated by molecular markers of drug effect, using
rubicin and methotrexate.45,48 Multilayered cell cultures are fluorescence-tagged antibodies that recognize cell proliferation
grown on collagen-coated microporous membranes: the rate (Ki67), antibodies that mark cell death or apoptosis (e.g., activated
of drug penetration can be evaluated by adding a drug on one caspase 3 or 6), and markers of DNA damage such as γH2AX.59
side of the MCC and measuring its concentration on the other We have validated the use of these biomarkers to study the distri-
as a function of time.51 This method has been used to demonstrate bution of activity of anticancer agents in solid tumors and have
slow tissue penetration of a range of clinically used chemothera- used quantitative IHC to demonstrate decreasing levels of activity
peutic agents, including etoposide, gemcitabine, paclitaxel, and with increasing distance from tumor blood vessels for doxorubi-
vinblastine46,52 (Fig. 4). cin, docetaxel,27,59 and paclitaxel (Figs. 5B, D).
Drug distribution can also be studied in animal models.
Growth of tumors in window chambers allows for direct observa-
tion of tumor microcirculation.53,54 Tumors can be excised at dif- Repopulation of Tumors Between Treatments
ferent times after drug treatment of animals bearing transplanted Repopulation of surviving cancer cells between daily frac-
tumors, including human tumor xenografts, and tissue sections tions during a course of radiotherapy has long been recognized
cut and processed for IHC. This analysis allows quantification as a cause of resistance. Similar repopulation occurs in the longer
of fluorescent drugs in relation to blood vessels or regions of hyp- intervals between courses of chemotherapy; in normal tissues, this
oxia (defined by a marker which identifies hypoxic cells, such allows recovery (e.g., of bone marrow), but in tumors, it may be a
as EF5 or pimonidazole), and the technique can be applied to cause of treatment failure.60,61 Our recent work has used dual la-
biopsies of human tumors.3,55 We have used this method to beling of hypoxic cells with EF5 and pimonidazole, with a vari-
quantify the distribution of the fluorescent drugs doxorubicin, able interval between applications of these markers, to determine
mitoxantrone, and topotecan in relation to blood vessels and the fate of tumor cells that were hypoxic at the time of treatment.

FIGURE 4. In vitro models to study drug distribution include spheroids and MCCs. The figure illustrates an MCC, which allows for a drug to be
added on one side and its concentration measured as a function of time on the other, thereby providing quantitative measurements of drug
penetration as a function of time.

© 2015 Wolters Kluwer Health, Inc. All rights reserved. www.journalppo.com 257

Copyright © 2015 Wolters Kluwer Health, Inc. All rights reserved.


Tan et al The Cancer Journal • Volume 21, Number 4, July/August 2015

FIGURE 5. Use of IHC to quantify drug distribution in model tumors. MCF7 tumors were treated with (A) doxorubicin (25 mg/kg
intravenously) or (B and C) paclitaxel (15 mg/kg intraperitoneally) or were untreated controls. A indicates doxorubicin fluorescence intensity
as a function of distance from the nearest blood vessel in tumor sections at 10 minutes after doxorubicin injection. B and C represent
percentage of positive pixels for biomarkers as a function of distance from the nearest blood vessel in the section. B. γH2AX at 10 minutes
after paclitaxel. C, Ki67 at 24 hours after paclitaxel. D, Photomicrographs of γH2AX at 10 minutes (left panels) and Ki67 at 24 hours (right
panels) in untreated (control) and paclitaxel-treated MCF7 xenografts.

For reasons discussed previously, hypoxic tumor cells are resistant evidence for improved therapeutic effects is minimal. Any strat-
to chemotherapy. We demonstrated that hypoxic cells that were egy that leads to decreased drug uptake in cells close to blood ves-
destined to die in the absence of treatment can survive and repop- sels will increase penetration to more distal cells; for example, we
ulate the tumor after chemotherapy62 have shown improved distribution of doxorubicin by inhibiting its
sequestration in acidic endosomes using PPIs,26 although the ef-
fect is modest and probably not the major mechanism by which
STRATEGIES TO IMPROVE THERAPY antitumor activity is increased by these agents.
Most studies of drug resistance have concentrated on the High IFP may have an adverse effect on treatment because it
multiple molecular causes of resistance in individual cancer may cause vascular compression and inadequate drug delivery
cells. Although these are important, even drug-sensitive cells through reduced convection. Human pancreatic tumors are ex-
can respond to treatment only if they are exposed to an ade- tremely resistant to systemic cancer therapy, and there is evidence
quate drug concentration. It is evident from the above that lim- that this is due to high IFP or to solid tissue stress due to elements
ited distribution within solid tumors limits exposure of tumor of the extracellular matrix preventing the delivery of drugs to
cells to many drugs, and limited drug distribution is an impor- constituent cells. Reduction of IFP using hyaluronidase or of
tant and neglected cause of drug resistance. A variety of strate- tumor-associated stroma tissue by other agents has been reported
gies are being researched to improve or complement the limited to improve delivery of chemotherapy in experimental models.65,66
distribution of commonly used anticancer drugs, and these are A recent study showed that imatinib, a molecular targeting drug,
described in the following sections. was able to reduce tumor IFP in melanoma and allowed delivery
of more doxorubicin into tumor tissue. Combined treatment
Improving Drug Distribution inhibited tumor growth and induced apoptosis of tumor cells.67
Several strategies might lead to improved drug distribution
within solid tumors. Giving drugs by continuous infusion to main-
tain levels in blood vessels can achieve this but will also increase Inhibiting Autophagy
delivery to normal tissues and will not necessarily improve the Agents that inhibit endosomal acidification, including (hy-
therapeutic index.63 Many investigators have delivered drugs in li- droxy)chloroquine and PPIs, can suppress autophagy, which is a
posomes or other nanoparticles, with the expectation that they potential survival mechanism following chemotherapy, especially
may selectively penetrate blood vessels in tumors and then release for nutrient-deprived cells. An Italian group reported the use of
drug over a prolonged period to increase tumor distribution,64 but PPIs to sensitize cancer cells to various chemotherapeutic agents.

258 www.journalppo.com © 2015 Wolters Kluwer Health, Inc. All rights reserved.

Copyright © 2015 Wolters Kluwer Health, Inc. All rights reserved.


The Cancer Journal • Volume 21, Number 4, July/August 2015 Drug Resistance Related to Solid Tumors

Multiple mechanisms are likely involved but appear to relate to Hypoxia-Activated Prodrugs
changes in acidity in both intracellular and extracellular compart- Hypoxia-activated prodrugs (HAPs) are administered in an
ments of tumor cells. Proton pump inhibitors can inhibit autoph- inactive form and are activated in the absence of oxygen. Hypoxic
agy probably because fusion of autophagosomes with acidic regions within solid tumors can be identified by IHC, using
endosomes is central to the process. We have confirmed that the markers of hypoxia such as pimonidazole and EF5. We have
PPI pantoprazole inhibits autophagy in vitro and in vivo and that shown that the HAP, TH-302, was able to increase expression of
it enhances activity of docetaxel against human tumor xenografts, γH2AX and cleaved caspases in hypoxic areas of human tumor
with improved distribution of activity, as determined by the bio- xenografts.71 The anticancer drugs docetaxel and doxorubicin
markers γH2AX and cleaved caspases throughout the tumors.27 had minimal activity in hypoxic regions, but the combination of
We showed marked effects of docetaxel and some other anticancer TH-302 and chemotherapy resulted in increased expression of
drugs to up-regulate autophagy as indicated by increased levels of γH2AX and cleaved caspases, compared with use of either drug
LC3B and reduced levels of p62 in all tumor regions (Fig. 6), with alone, in regions both proximal and distal to blood vessels. Thus,
opposite effects indicating inhibition of autophagy, when chemo- the distributions of activity of the chemotherapy drugs and of the
therapy is combined with pantoprazole. We obtained further evi- HAP TH-302 complement each other, although there appear to be
dence for this being the main mechanism of action by using additional effects to enhance overall activity.62 The combination
autophagy-deficient cells generated by shRNA knockdown of of gemcitabine and TH-302 has given encouraging results in a
the autophagy proteins ATG7 and BECLIN1 or both.27 Several phase II trial for human pancreatic cancer,72 and a phase III trial
other studies have shown that PPIs such as omeprazole, is in progress.
esomeprazole, and pantoprazole have activity against human he-
matopoietic and solid tumors; they may revert chemoresistance
in drug-resistant tumors and directly induce killing of tumor Inhibition of Tumor Cell Repopulation
cells.24,68,69 Our unpublished data show that a wide spectrum of Therapeutic index might be improved by inhibiting selec-
anticancer drugs up-regulate autophagy in cultured tumor cells tively the repopulation of tumor cells that have the ability to regen-
and that this can be inhibited by PPIs. Growing evidence suggests erate the tumor between courses of chemotherapy, without
that the major mechanism by which PPIs enhance the effects of inhibiting repopulation of critical normal tissues. This has been
chemotherapy is by inhibition of autophagy27,70 attempted in model systems using cytostatic inhibitors such as

FIGURE 6. Demonstration of the effect of drugs on the autophagy markers LC3 (indicating increased autophagy) and p62 (indicating
decreased autophagy) in different regions of PC3 human prostate cancer xenografts. Tumors treated with docetaxel (15 mg/kg
intraperitoneally), pantoprazole (200 mg/kg intraperitoneally), pantoprazole 2 hours prior to docetaxel, or untreated controls. Figures
represent percentage of positive pixels for LC3 (A, C) or p62 (B, D) as a function of distance from the nearest blood vessel (A, B) or as a
function of distance from the nearest hypoxic region (C, D). Note that docetaxel increases autophagy (increased LC3, decreased p62)
throughout the tumors and that this is inhibited by pantoprazole. *Figure adapted from Tan et al.27

© 2015 Wolters Kluwer Health, Inc. All rights reserved. www.journalppo.com 259

Copyright © 2015 Wolters Kluwer Health, Inc. All rights reserved.


Tan et al The Cancer Journal • Volume 21, Number 4, July/August 2015

FIGURE 7. The figure demonstrates the fate of cells in prostate cancer PC3 xenografts that were initially hypoxic, as indicated by labeling with
pimonidazole, and were then exposed to a second hypoxia marker (EF5) 24 hours later. A shows the percentage of originally hypoxic cells
(pimo+) that are still present in the tumor and have reoxygenated (i.e., are pimo+, EF5−) in control tumors and in tumors at 24 hours after
treatment with docetaxel (DOC), TH-302, or the combination. Note that docetaxel alone induced the highest rate of reoxygenation and
that this is inhibited by TH-302. B indicates Ki67 staining (a marker of proliferation) in the reoxygenated cells, which is also highest after
chemotherapy and inhibited by TH-302. C indicates Ki67 staining in relation to the nearest hypoxic area and demonstrates that docetaxel
alone leads to increased Ki67 levels compared with controls and combination therapy in regions close to hypoxia. *Figure adapted from
Saggar and Tannock.62

hormonal agents in breast cancer and inhibitors of growth fac- ACKNOWLEDGMENTS


tors,73,74 but effects on outcome have been modest. As indicated The authors thank all members of the Pathology Research
above, the poor distribution of drugs in solid tumors leads to sur- Program (PRP) and the Advanced Optical Microscopy Facility.
vival of hypoxic and other poorly nourished cells, which might
then repopulate the tumor. By administering pimonidazole and REFERENCES
EF5 sequentially and labeling with the proliferation marker
1. Minchinton AI, Tannock IF. Drug penetration in solid tumours. Nat Rev
Ki67, we have been able to follow reoxygenation and proliferation
Cancer. 2006;6:583–592.
of tumor cells that were hypoxic at the time of administration of
2. Tredan O, Galmarini CM, Patel K, et al. Drug resistance and the solid tumor
chemotherapy.62 We demonstrated that chemotherapy (with doxo- microenvironment. J Natl Cancer Inst. 2007;99:1441–1454.
rubicin or docetaxel) induced repopulation and reoxygenation of 3. Lankelma J, Dekker H, Luque FR, et al. Doxorubicin gradients in human
tumor cells that were previously hypoxic. Moreover, these pro- breast cancer. Clin Cancer Res. 1999;5:1703–1707.
cesses were inhibited by the HAP TH-302, indicating another po- 4. Huxham LA, Kyle AH, Baker JH, et al. Microregional effects of
tential mechanism whereby combined targeting of well-nourished gemcitabine in HCT-116 xenografts. Cancer Res. 2004;64:6537–6541.
cells with chemotherapy, and hypoxic cells with HAPs, might im- 5. Hirst DG, Denekamp J. Tumour cell proliferation in relation to the vascula-
prove therapeutic effects62 (Fig. 7). ture. Cell Tissue Kinet. 1979;12:31–42.
6. Ljungkvist AS, Bussink J, Rijken PF, et al. Vascular architecture, hypoxia,
and proliferation in first-generation xenografts of human head-and-neck
squamous cell carcinomas. Int J Radiat Oncol Biol Phys. 2002;54:
CONCLUSIONS
215–228.
The effectiveness of anticancer drugs is impaired by their 7. Vaupel P. Tumor microenvironmental physiology and its implications for
limited distribution within tumors. Agents that modify or comple- radiation oncology. Semin Radiat Oncol. 2004;14:198–206.
ment the activity of anticancer drugs have the ability to improve 8. Vaupel P, Harrison L. Tumor hypoxia: causative factors, compensatory
treatment outcomes. Modification of autophagy to increase the mechanisms, and cellular response. Oncologist. 2004;9(suppl 5):4–9.
toxic activity of drugs within tumors and use of hypoxia- 9. Chaplin DJ, Trotter MJ, Durand RE, et al. Evidence for intermittent radio-
activated prodrugs hold particular promise to improve the effec- biological hypoxia in experimental tumour systems. Biomed Biochim Acta.
tiveness of conventional chemotherapy. 1989;48:S255–S259.

260 www.journalppo.com © 2015 Wolters Kluwer Health, Inc. All rights reserved.

Copyright © 2015 Wolters Kluwer Health, Inc. All rights reserved.


The Cancer Journal • Volume 21, Number 4, July/August 2015 Drug Resistance Related to Solid Tumors

10. Durand RE, LePard NE. Contribution of transient blood flow to tumour 34. Klionsky DJ, Abdalla FC, Abeliovich H, et al. Guidelines for the use and
hypoxia in mice. Acta Oncol. 1995;34:317–323. interpretation of assays for monitoring autophagy. Autophagy. 2012;8:
11. Graeber TG, Osmanian C, Jacks T, et al. Hypoxia-mediated selection of 445–544.
cells with diminished apoptotic potential in solid tumours. Nature. 1996; 35. Bjorkoy G, Lamark T, Pankiv S, et al. Monitoring autophagic degradation
379:88–91. of p62/SQSTM1. Methods Enzymol. 2009;452:181–197.
12. Haensgen G, Krause U, Becker A, et al. Tumor hypoxia, p53, and prognosis 36. Li J, Hou N, Faried A, et al. Inhibition of autophagy by 3-MA enhances the
in cervical cancers. Int J Radiat Oncol Biol Phys. 2001;50:865–872. effect of 5-FU–induced apoptosis in colon cancer cells. Ann Surg Oncol.
13. Ogiso Y, Tomida A, Lei S, et al. Proteasome inhibition circumvents solid 2009;16:761–771.
tumor resistance to topoisomerase II–directed drugs. Cancer Res. 2000; 37. Karpathiou G, Sivridis E, Koukourakis MI, et al. Light-chain 3A autopha-
60:2429–2434. gic activity and prognostic significance in non–small cell lung carcinomas.
14. Wartenberg M, Ling FC, Müschen M, et al. Regulation of the multidrug re- Chest. 2011;140:127–134.
sistance transporter P-glycoprotein in multicellular tumor spheroids by 38. Sivridis E, Koukourakis MI, Mendrinos SE, et al. Beclin-1 and LC3A ex-
hypoxia-inducible factor (HIF-1) and reactive oxygen species. FASEB pression in cutaneous malignant melanomas: a biphasic survival pattern
J. 2003;17:503–505 for beclin-1. Melanoma Res. 2011;21:188–195.
15. Azad MB, Chen Y, Henson ES, et al. Hypoxia induces autophagic cell 39. Sharma N, Thomas S, Golden EB, et al. Inhibition of autophagy and induc-
death in apoptosis-competent cells through a mechanism involving BNIP3. tion of breast cancer cell death by mefloquine, an antimalarial agent. Can-
Autophagy. 2008;4:195–204. cer Lett. 2012;326:143–154.
16. Rouschop KM, van den Beucken T, Dubois L, et al. The unfolded protein 40. Yang ZJ, Chee CE, Huang S, et al. The role of autophagy in cancer: thera-
response protects human tumor cells during hypoxia through regulation of peutic implications. Mol Cancer Ther. 2011;10:1533–1541.
the autophagy genes MAP1LC3B and ATG5. J Clin Invest. 2010;120: 41. Jain RK, Baxter LT. Mechanisms of heterogeneous distribution of mono-
127–141. clonal antibodies and other macromolecules in tumors: significance of ele-
17. Warburg O. On the origin of cancer cells. Science. 1956;123:309–314. vated interstitial pressure. Cancer Res. 1988;48(24 pt 1):7022–7032.
18. Tannock IF, Rotin D. Acid pH in tumors and its potential for therapeutic ex- 42. Jain RK. Delivery of novel therapeutic agents in tumors: physiological bar-
ploitation. Cancer Res. 1989;49:4373–4384. riers and strategies. J Natl Cancer Inst. 1989;81:570–576.
19. Svastova E, Hulíková A, Rafajová M, et al. Hypoxia activates the capacity 43. Undevia SD, Gomez-Abuin G, Ratain MJ. Pharmacokinetic variability of
of tumor-associated carbonic anhydrase IX to acidify extracellular pH. anticancer agents. Nat Rev Cancer. 2005;5:447–458.
FEBS Lett. 2004;577:439–445. 44. Conger AD, Ziskin MC. Growth of mammalian multicellular tumor spher-
20. Huang Z, Huang Y. The change of intracellular pH is involved in the oids. Cancer Res. 1983;43:556–560.
cisplatin-resistance of human lung adenocarcinoma A549/DDP cells. Can- 45. Sutherland RM. Cell and environment interactions in tumor microregions:
cer Invest. 2005;23:26–32. the multicell spheroid model. Science. 1988;240:177–184.
21. Patel KJ, Trédan O, Tannock IF. Distribution of the anticancer drugs doxo- 46. Tannock IF, Lee CM, Tunggal JK, et al. Limited penetration of anticancer
rubicin, mitoxantrone and topotecan in tumors and normal tissues. Cancer drugs through tumor tissue: a potential cause of resistance of solid tumors
Chemother Phamacol. in press;72:127–138. to chemotherapy. Clin Cancer Res. 2002;8:878–884.
22. Lee CM, Tannock IF. Inhibition of endosomal sequestration of basic anti- 47. Hicks KO, Pruijn FB, Secomb TW, et al. Use of three-dimensional tissue cul-
cancer drugs: influence on cytotoxicity and tissue penetration. Br J Cancer. tures to model extravascular transport and predict in vivo activity of hypoxia-
2006;94:863–869. targeted anticancer drugs. J Natl Cancer Inst. 2006;98:1118–1128.
23. Luciani F, Spada M, De Milito A, et al. Effect of proton pump inhibitor pre- 48. Durand RE. Distribution and activity of antineoplastic drugs in a tumor
treatment on resistance of solid tumors to cytotoxic drugs. J Natl Cancer model. J Natl Cancer Inst. 1989;81:146–152.
Inst. 2004;96:1702–1713. 49. Lesser GJ, Grossman SA, Eller S, et al. The distribution of systemically ad-
24. De Milito A, Iessi E, Logozzi M, et al. Proton pump inhibitors induce ministered [3H]-paclitaxel in rats: a quantitative autoradiographic study.
apoptosis of human B-cell tumors through a caspase-independent Cancer Chemother Pharmacol. 1995;37:73–78.
mechanism involving reactive oxygen species. Cancer Res. 2007;67: 50. Kuh HJ, Jang SH, Wientjes MG, et al. Determinants of paclitaxel penetra-
5408–5417. tion and accumulation in human solid tumor. J Pharmacol Exp Ther. 1999;
25. Marino ML, Fais S, Djavaheri-Mergny M, et al. Proton pump inhibition in- 290:871–880.
duces autophagy as a survival mechanism following oxidative stress in hu- 51. Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nat Rev Cancer.
man melanoma cells. Cell Death Dis. 2010;1:e87. 2011;11:393–410.
26. Patel KJ, Lee C, Tan Q, et al. Use of the proton pump inhibitor pantoprazole 52. Choi MS, Kim SH, Kuh HJ. Penetration of paclitaxel and 5-fluorouracil in
to modify the distribution and activity of doxorubicin: a potential strategy multicellular layers of human colorectal cancer cells. Oncol Rep. 2011;25:
to improve the therapy of solid tumors. Clin Cancer Res. 2013;19: 863–870.
6766–6776. 53. Yuan F, Salehi HA, Boucher Y, et al. Vascular permeability and microcircu-
27. Tan Q, Joshua AM, Saggar JK, et al. Effect of pantoprazole to enhance ac- lation of gliomas and mammary carcinomas transplanted in rat and mouse
tivity of docetaxel against human tumour xenografts by inhibiting autoph- cranial windows. Cancer Res. 1994;54:4564–4568.
agy. Br J Cancer. 2015;112:832–840. 54. Helmlinger G, Yuan F, Dellian M, et al. Interstitial pH and PO2 gradients in
28. Hoyer-Hansen M, Jaattela M. Connecting endoplasmic reticulum stress to solid tumors in vivo: high-resolution measurements reveal a lack of corre-
autophagy by unfolded protein response and calcium. Cell Death Differ. lation. Nat Med. 1997;3:177–182.
2007;14:1576–1582. 55. Brana I, Ocana A, Chen EX, et al. A phase I trial of pantoprazole in com-
29. Mizushima N, Ohsumi Y, Yoshimori T. Autophagosome formation in bination with doxorubicin in patients with advanced solid tumors: evalua-
mammalian cells. Cell Struct Funct. 2002;27:421–429. tion of pharmacokinetics of both drugs and tissue penetration of
30. Gozuacik D, Kimchi A. Autophagy as a cell death and tumor suppressor doxorubicin. Invest New Drugs. 2014;32:1269–1277.
mechanism. Oncogene. 2004;23:2891–2906. 56. Primeau AJ, Rendon A, Hedley D, et al. The distribution of the anticancer
31. Ganley IG, Wong PM, Gammoh N, et al. Distinct autophagosomal- drug doxorubicin in relation to blood vessels in solid tumors. Clin Cancer
lysosomal fusion mechanism revealed by thapsigargin-induced autophagy Res. 2005;11:8782–8788.
arrest. Mol Cell. 2011;42:731–743. 57. Patel KJ, Tredan O, Tannock IF. Distribution of the anticancer drugs doxo-
32. Tsukada M, Ohsumi Y. Isolation and characterization of autophagy- rubicin, mitoxantrone and topotecan in tumors and normal tissues. Cancer
defective mutants of Saccharomyces cerevisiae. FEBS Lett. 1993;333: Chemother Pharmacol. 2013;72:127–138.
169–174. 58. Lee CM, Tannock IF. The distribution of the therapeutic monoclonal anti-
33. Tanida I, Ueno T, Kominami E. LC3 conjugation system in mammalian au- bodies cetuximab and trastuzumab within solid tumors. BMC Cancer.
tophagy. Int J Biochem Cell Biol. 2004;36:2503–2518. 2010;10:255.

© 2015 Wolters Kluwer Health, Inc. All rights reserved. www.journalppo.com 261

Copyright © 2015 Wolters Kluwer Health, Inc. All rights reserved.


Tan et al The Cancer Journal • Volume 21, Number 4, July/August 2015

59. Saggar JK, Fung AS, Patel KJ, et al. Use of molecular biomarkers to quan- 67. Fan Y, Du W, He B, et al. The reduction of tumor interstitial fluid pressure
tify the spatial distribution of effects of anticancer drugs in solid tumors. by liposomal imatinib and its effect on combination therapy with liposomal
Mol Cancer Ther. 2013;12:542–552. doxorubicin. Biomaterials. 2013;34:2277–2288.
60. Tannock IF. Tumor physiology and drug resistance. Cancer Metastasis Rev. 68. Yeo M, Kim DK, Kim YB, et al. Selective induction of apoptosis with pro-
2001;20:123–132. ton pump inhibitor in gastric cancer cells. Clin Cancer Res. 2004;10:
61. Kim JJ, Tannock IF. Repopulation of cancer cells during therapy: an impor- 8687–8696.
tant cause of treatment failure. Nat Rev Cancer. 2005;5:516–525. 69. De Milito A, Canese R, Marino ML, et al. pH-dependent antitumor activity
62. Saggar JK, Tannock IF. Chemotherapy rescues hypoxic tumor cells and in- of proton pump inhibitors against human melanoma is mediated by inhibi-
duces their reoxygenation and repopulation—an effect that is inhibited tion of tumor acidity. Int J Cancer. 2010;127:207–219.
by the hypoxia-activated pro-drug TH-302. Clin Cancer Res. 2015;21: 70. Udelnow A, Kreyes A, Ellinger S, et al. Omeprazole inhibits proliferation and
2107–2114. modulates autophagy in pancreatic cancer cells. PLoS One. 2011;6:e20143.
63. Tahover E, Patil YP, Gabizon AA. Emerging delivery systems to reduce 71. Saggar JK, Tannock IF. Activity of the hypoxia-activated pro-drug TH-302
doxorubicin cardiotoxicity and improve therapeutic index: focus on lipo- in hypoxic and perivascular regions of solid tumors and its potential to en-
somes. Anticancer Drugs. 2015;26:241–258. hance therapeutic effects of chemotherapy. Int J Cancer. 2014;134:
64. Vippola M, Falck GC, Lindberg HK, et al. Preparation of nanoparticle 2726–2734.
dispersions for in-vitro toxicity testing. Hum Exp Toxicol. 2009;28: 72. Borad MJ, Reddy SG, Bahary N, et al. Randomized phase II trial of
377–385. gemcitabine plus TH-302 versus gemcitabine in patients with advanced
65. Olive KP, Jacobetz MA, Davidson CJ, et al. Inhibition of Hedgehog signal- pancreatic cancer. J Clin Oncol. 2015;33:1475–1481.
ing enhances delivery of chemotherapy in a mouse model of pancreatic can- 73. Davis AJ, Tannock JF. Repopulation of tumour cells between cycles of che-
cer. Science. 2009;324:1457–1461. motherapy: a neglected factor. Lancet Oncol. 2000;1:86–93.
66. Provenzano PP, Cuevas C, Chang AE, et al. Enzymatic targeting of the 74. Wu L, Tannock IF. Selective estrogen receptor modulators as inhibitors of
stroma ablates physical barriers to treatment of pancreatic ductal adenocar- repopulation of human breast cancer cell lines after chemotherapy. Clin
cinoma. Cancer Cell. 2012;21:418–429. Cancer Res. 2003;9:4614–4618.

262 www.journalppo.com © 2015 Wolters Kluwer Health, Inc. All rights reserved.

Copyright © 2015 Wolters Kluwer Health, Inc. All rights reserved.

You might also like