Jump to content

Antineoplastic resistance: Difference between revisions

From Wikipedia, the free encyclopedia
Content deleted Content added
mNo edit summary
Xiazcy (talk | contribs)
An major overhaul of the old page, with the addition of information in over 2000 words
Line 1: Line 1:
'''Antineoplastic resistance''' is the [[multidrug resistance]] of neoplastic ([[cancer]]ous) cells, rather than drug resistance involving microorganisms such as [[bacteria]], fungi and viruses. Cancer cells have the ability to become resistant to multiple drugs by many mechanisms:<ref>Resistance to cancer chemotherapy: failure in drug response from ADME to P-gp. http://www.cancerci.com/content/15/1/71</ref>
'''Antineoplastic resistance''', often used interchangeably with chemotherapy resistance, is the multiple drug resistance of neoplastic (cancerous) cells, or the ability of cancer cells to survive and grow despite anti-cancer therapies.


There are two general causes of antineoplastic therapy failure<ref name=":0">{{Cite journal
*Increased efflux of drug (as by [[P-glycoprotein]] (ABCB1), [[multidrug resistance-associated protein]] (ABCC1), and breast cancer resistance protein (also known as mitoxantrone resistance associated protein, MXR, or [[ABCG2]])
| last = Luqmani
*[[Antibiotic resistance#Mechanisms|Enzymatic deactivation]] (i.e., glutathione conjugation)
| first = Y.A.
*Decreased permeability (drugs cannot enter the cell)
| title = Mechanisms of Drug Resistance in Cancer Chemotherapy
*Altered [[Binding site|binding-sites]]
| url = http://www.karger.com/doi/10.1159/000086183
*Alternate [[metabolic pathways]] (the cancer compensates for the effect of the drug).
| journal = Medical Principles and Practice
*Gene amplification/genomic instability
| volume = 14
| issue = 1
| pages = 35–48
| doi = 10.1159/000086183
}}</ref>:
# Possession of inherent properties, such as genetic characteristics, which gave cancer cells their resistance<ref name=":0" />. Such idea is rooted in the concept of [[Tumour heterogeneity|cancer cell heterogeneity]].
# Acquired resistance after drug exposure<ref name=":0" />. Cancer cells have the ability to become resistant to multiple drugs by various mechanisms<ref name=":0" /> <ref name=":1">{{Cite journal
| last = Housman
| first = Genevieve
| last2 = Byler
| first2 = Shannon
| last3 = Heerboth
| first3 = Sarah
| last4 = Lapinska
| first4 = Karolina
| last5 = Longacre
| first5 = Mckenna
| last6 = Snyder
| first6 = Nicole
| last7 = Sarkar
| first7 = Sibaji
| date = 2014-09-05
| title = Drug Resistance in Cancer: An Overview
| url = http://www.mdpi.com/2072-6694/6/3/1769
| journal = Cancers
| language = en
| volume = 6
| issue = 3
| pages = 1769–1792
| doi = 10.3390/cancers6031769
| pmc = 4190567
| pmid = 25198391
}}</ref>
#* Altered membrane transport
#* Enhanced DNA repair
#* Apoptotic pathway defects
#* Alteration of target molecules
#* Protein and pathway mechanisms such as enzymatic deactivation
As cancer is very much a disease of genetic mutations, underlying the development of acquired drug resistance mechanisms are several genomic events.
* Genome alterations (e.g. gene amplification and deletion)
* Epigenetic modifications
The mechanisms of antineoplastic resistance is in no way exhaustive. Cancer cells ultimately employ a variety of tools, involving genes, proteins and altered pathways, to ensure their survival against drugs. As such, the treatment of cancer is a very complex and challenging field.


== Cancer Cell Heterogeneity ==
Because efflux is a significant contributor for multidrug resistance in cancer cells, research has been aimed at blocking specific efflux mechanisms.<ref name="pmid16690355">{{cite journal | author = Modok S, Mellor HR, Callaghan R | title = Modulation of multidrug resistance efflux pump activity to overcome chemoresistance in cancer | journal = Curr Opin Pharmacol | volume = 6 | issue = 4 | pages = 350–4 |date=August 2006 | pmid = 16690355 | doi = 10.1016/j.coph.2006.01.009 }}</ref> Treatment of cancer is complicated by the fact that there is a variety of different DNA mutations that cause or contribute to tumor formation, as well as myriad mechanisms by which cells resist drugs.
Cancer cell heterogeneity, or [[tumour heterogeneity]] is the idea that, within a tumor mass, a certain population of cancer cells exists that possess inherent genetic characteristics that confer drug resistance. The administration of antineoplastic drugs acts as a selection mechanism that kills non-resistant cells, while favoring these resistant cells. While the initial tumor mass may shrink as an initial response to the drug, these resistant colonies will survive treatment and then propagate, eventually causing a cancer relapse.


A slightly different way in which cancer cell heterogeneity gives rise to disease progression is via [[Targeted therapy|targeted therapies]], which target a specific molecular marker. Tumor cells that do not express the specific marker are not killed, and are then able to divide and mutate further, creating a new heterogeneous tumor.
Notable differences between antibiotic drugs and antineoplastic (anticancer) drugs that complicate their design are that cancer cells are altered human cells and thus more difficult to damage without damaging healthy cells.


Nonetheless, the presence of such genomic heterogeneity complicates antineoplastic treatment.
==See also==
[[File:Antineoplastic resistances.png|thumb|440x440px|An overview of antineoplastic resistance mechanisms]]
*[[Antineoplastic drugs]]
*[[Drug resistance]]


== Mechanisms of Acquired Resistance ==
==References==
{{Reflist}}


=== Alteration of Membrane Transport ===
==Further reading==
Many classes of antineoplastic drugs acts on DNA, nuclear components, and intracellular pathways, meaning that they require entry into the cancer cells. The [[p-glycoprotein]] (P-gp), or the multiple drug resistance protein, is a phosphorylated and glycosylated membrane transporter that can shuttle drugs out of the cell, thereby decreasing or ablating drug efficacy. This transporter protein is encoded by the ''[[P-glycoprotein|MDR1]]'' gene and is also called the ATP-binding cassette (ABC) protein.  MDR1 has promiscuous substrate specificity, allowing it to be able to transport many structurally diverse compounds across the cell membrane, mainly hydrophobic compounds. Studies have found that the MDR1 gene can be activated and overexpressed in response to drug administration, thus forming the basis for resistance to many drugs<ref name=":0" />. Overexpression of the MDR1 gene in cancer cells is used to keep intracellular levels of antineoplastic drugs below cell-killing levels.
* {{cite journal | author = Guminski AD, Harnett PR, deFazio A | title = Scientists and clinicians test their metal-back to the future with platinum compounds | journal = Lancet Oncol. | volume = 3 | issue = 5 | pages = 312–8 |date=May 2002 | pmid = 12067809 | doi = 10.1016/S1470-2045(02)00733-7 }}
* {{cite journal | author = Krishan A | title = Monitoring of cellular resistance to cancer chemotherapy: drug retention and efflux | journal = Methods Cell Biol. | volume = 64 | issue = | pages = 193–209 | year = 2001 | pmid = 11070840 | doi = 10.1016/s0091-679x(01)64014-7}}
{{refend}}


For example, rifampicin has been found to induce ''MDR1'' expression. Experiments in differe
== External links==

* [http://crdd.osdd.net/raghava/cancerdr/ CancerDR]: Cancer Drug Resistance Database. [http://www.nature.com/srep/2013/130313/srep01445/full/srep01445.html Scientific Reports 3, 1445]
nt drug resistant cell lines and patient DNA revealed gene rearrangements that had initiated the activation or overexpression of ''MDR1''<ref>{{Cite journal
| last = Mickley
| first = L A
| last2 = Spengler
| first2 = B A
| last3 = Knutsen
| first3 = T A
| last4 = Biedler
| first4 = J L
| last5 = Fojo
| first5 = T
| date = 1997-04-15
| title = Gene rearrangement: a novel mechanism for MDR-1 gene activation.
| url = http://www.jci.org/articles/view/119362
| journal = Journal of Clinical Investigation
| language = en
| volume = 99
| issue = 8
| pages = 1947–1957
| doi = 10.1172/jci119362
}}</ref>. A C3435T polymorphism in exon 226 of ''MDR1'' has also been strongly correlated with p-glycoprotein activities<ref>{{Cite journal
| last = Hoffmeyer
| first = S.
| last2 = Burk
| first2 = O.
| last3 = Richter
| first3 = O. von
| last4 = Arnold
| first4 = H. P.
| last5 = Brockmöller
| first5 = J.
| last6 = Johne
| first6 = A.
| last7 = Cascorbi
| first7 = I.
| last8 = Gerloff
| first8 = T.
| last9 = Roots
| first9 = I.
| date = 2000-03-28
| title = Functional polymorphisms of the human multidrug-resistance gene: Multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo
| url = http://www.pnas.org/content/97/7/3473
| journal = Proceedings of the National Academy of Sciences
| language = en
| volume = 97
| issue = 7
| pages = 3473–3478
| doi = 10.1073/pnas.97.7.3473
| issn = 0027-8424
}}</ref>.

Studies have shown that ''MDR1'' activation occurs through activation of [[NF-κB]], a protein complex that acts as a transcription factor<ref>{{Cite journal
| last = Bentires-Alj
| first = Mohamed
| last2 = Barbu
| first2 = Veronique
| last3 = Fillet
| first3 = Marianne
| last4 = Chariot
| first4 = Alain
| last5 = Relic
| first5 = Biserka
| last6 = Jacobs
| first6 = Nathalie
| last7 = Gielen
| first7 = Jacques
| last8 = Merville
| first8 = Marie-Paule
| last9 = Bours
| first9 = Vincent
| date = 2003-01-01
| title = NF-κB transcription factor induces drug resistance through MDR1 expression in cancer cells
| url = http://www.nature.com/onc/journal/v22/n1/full/1206056a.html
| journal = Oncogene
| language = en
| volume = 22
| issue = 1
| pages = 90–97
| doi = 10.1038/sj.onc.1206056
| issn = 0950-9232
}}</ref><ref>{{Cite journal
| last = Kim
| first = Hyung Gyun
| last2 = Hien
| first2 = Tran Thi
| last3 = Han
| first3 = Eun Hee
| last4 = Hwang
| first4 = Yong Pil
| last5 = Choi
| first5 = Jae Ho
| last6 = Kang
| first6 = Keon Wook
| last7 = Kwon
| first7 = Kwang-il
| last8 = Kim
| first8 = Bong-Hee
| last9 = Kim
| first9 = Sang Kyum
| date = 2011-03-01
| title = Metformin inhibits P-glycoprotein expression via the NF-κB pathway and CRE transcriptional activity through AMPK activation
| url = http://onlinelibrary.wiley.com/doi/10.1111/j.1476-5381.2010.01101.x/abstract
| journal = British Journal of Pharmacology
| language = en
| volume = 162
| issue = 5
| pages = 1096–1108
| doi = 10.1111/j.1476-5381.2010.01101.x
| issn = 1476-5381
| pmc = 3051382
| pmid = 21054339
}}</ref>. There is a NF-κB binding site located adjacent to the rat mdr1b gene<ref>{{Cite journal
| last = Zhou
| first = Ge
| last2 = Kuo
| first2 = M. Tien
| date = 1997-06-13
| title = NF-κB-mediated Induction of mdr1b Expression by Insulin in Rat Hepatoma Cells
| url = http://www.jbc.org/content/272/24/15174
| journal = Journal of Biological Chemistry
| language = en
| volume = 272
| issue = 24
| pages = 15174–15183
| doi = 10.1074/jbc.272.24.15174
| issn = 0021-9258
}}</ref>, and studies show that MDR1 upregulation is reliant on NF-κB activation as well<ref>{{Cite journal
| last = Thévenod
| first = Frank
| last2 = Friedmann
| first2 = Jenny M.
| last3 = Katsen
| first3 = Alice D.
| last4 = Hauser
| first4 = Ingeborg A.
| date = 2000-01-21
| title = Up-regulation of Multidrug Resistance P-glycoprotein via Nuclear Factor-κB Activation Protects Kidney Proximal Tubule Cells from Cadmium- and Reactive Oxygen Species-induced Apoptosis
| url = http://www.jbc.org/content/275/3/1887
| journal = Journal of Biological Chemistry
| language = en
| volume = 275
| issue = 3
| pages = 1887–1896
| doi = 10.1074/jbc.275.3.1887
| issn = 0021-9258
}}</ref><ref>{{Cite journal
| last = Kuo
| first = Macus Tien
| last2 = Liu
| first2 = Zesheng
| last3 = Wei
| first3 = Yingjie
| last4 = Lin-Lee
| first4 = Yen-chiu
| last5 = Tatebe
| first5 = Shigeru
| last6 = Mills
| first6 = Gordon B.
| last7 = Unate
| first7 = Hitoshi
| date = 2002-03-27
| title = Induction of human MDR1 gene expression by 2-acetylaminofluorene is mediated by effectors of the phosphoinositide 3-kinase pathway that activate NF-kappaB signaling
| url = http://www.ncbi.nlm.nih.gov/pubmed/11960367
| journal = Oncogene
| volume = 21
| issue = 13
| pages = 1945–1954
| doi = 10.1038/sj.onc.1205117
| issn = 0950-9232
| pmid = 11960367
}}</ref>. NF-κB is active in tumour cells due to mutations in the NF-κB gene or in the inhibitory IκB gene. In [[Colorectal cancer|colon cancer]] cells, inhibition of NF-κB or MDR1 resulted in increased [[apoptosis]] in response to a chemotherapeutic agent used to treat various cancers<ref>{{Cite journal
| last = Bentires-Alj
| first = Mohamed
| last2 = Barbu
| first2 = Veronique
| last3 = Fillet
| first3 = Marianne
| last4 = Chariot
| first4 = Alain
| last5 = Relic
| first5 = Biserka
| last6 = Jacobs
| first6 = Nathalie
| last7 = Gielen
| first7 = Jacques
| last8 = Merville
| first8 = Marie-Paule
| last9 = Bours
| first9 = Vincent
| date = 2003-01-01
| title = NF-κB transcription factor induces drug resistance through MDR1 expression in cancer cells
| url = http://www.nature.com/onc/journal/v22/n1/full/1206056a.html
| journal = Oncogene
| language = en
| volume = 22
| issue = 1
| pages = 90–97
| doi = 10.1038/sj.onc.1206056
| issn = 0950-9232
}}</ref>.

=== Enhanced DNA Repair ===
Enhanced [[DNA repair]] plays an important role in the ability for cancer cells to overcome drug-induced DNA damages.

Platinum-base chemotherapy, such as [[cisplatin]], targets tumor cells by cross-linking their DNA strands, causing mutation and damage<ref name=":0" />. Such damage will trigger programmed cell death ([[apoptosis]]) in cancer cells. Cisplatin resistance occurs when cancer cells develop an enhanced ability to reverse such damage, by removing the cisplatin from DNA and repairing any damage done<ref name=":0" /><ref name=":1" />. The unregulated expression of the excision repair cross-complementing ([[ERCC1]]) gene and protein in cisplatin-resistant tumors underlies such process<ref name=":0" />.

Some chemotherapies are [[alkylating agents]] that result in the attachment of an alkyl group to DNA. O6-methylguanine DNA methyltransferase (MGMT) is a DNA repair enzyme that removes alkyl groups from DNA. Expression of ''MGMT'' is upregulated in many cancer cells, providing a protective function from alkylating agents<ref name=":1" />. Increased MGMT expression has been found in colon cancer, lung cancer, non-Hodgkin’s lymphoma, breast cancer, gliomas, myeloma and pancreatic cancer<ref>{{Cite journal
| last = Gerson
| first = Stanton L.
| title = MGMT: its role in cancer aetiology and cancer therapeutics
| url = http://www.nature.com/doifinder/10.1038/nrc1319
| journal = Nature Reviews Cancer
| volume = 4
| issue = 4
| pages = 296–307
| doi = 10.1038/nrc1319
}}</ref>.

=== Apoptotic Pathway Defects ===
''[[P53|TP53]]'' is a [[tumor suppressor gene]] that encodes the p53 protein. It is a protein that responds to DNA damage, either by initiating [[DNA repair]], cell cycle arrest, or [[apoptosis]]. The loss of ''TP53'' via [[Deletion (genetics)|gene deletion]] can result in continued replication despite DNA damage. This tolerance of DNA damage can grant cancer cells a method of resistance to drugs aiming to induce apoptosis via DNA damage<ref name=":0" /><ref name=":1" />.

Other genes involved in the apoptotic pathway related drug resistance includes ''[[HRAS|h-ras]]'' and ''bcl-2/bax''<ref>{{Cite journal
| last = Burger
| first = H.
| last2 = Nooter
| first2 = K.
| last3 = Boersma
| first3 = A. W.
| last4 = van Wingerden
| first4 = K. E.
| last5 = Looijenga
| first5 = L. H.
| last6 = Jochemsen
| first6 = A. G.
| last7 = Stoter
| first7 = G.
| date = 1999-05-17
| title = Distinct p53-independent apoptotic cell death signalling pathways in testicular germ cell tumour cell lines
| url = http://www.ncbi.nlm.nih.gov/pubmed/10225454
| journal = International Journal of Cancer. Journal International Du Cancer
| volume = 81
| issue = 4
| pages = 620–628
| issn = 0020-7136
| pmid = 10225454
}}</ref>. Oncogenic ''h-ras'' has been found to increase expression of ERCC1, resulting in enhanced DNA repair (see above)<ref>{{Cite journal
| last = Youn
| first = Cha-Kyung
| last2 = Kim
| first2 = Mi-Hwa
| last3 = Cho
| first3 = Hyun-Ju
| last4 = Kim
| first4 = Hong-Beum
| last5 = Chang
| first5 = In-Youb
| last6 = Chung
| first6 = Myung-Hee
| last7 = You
| first7 = Ho Jin
| date = 2004-07-15
| title = Oncogenic H-Ras Up-Regulates Expression of ERCC1 to Protect Cells from Platinum-Based Anticancer Agents
| url = http://cancerres.aacrjournals.org/content/64/14/4849
| journal = Cancer Research
| language = en
| volume = 64
| issue = 14
| pages = 4849–4857
| doi = 10.1158/0008-5472.CAN-04-0348
| issn = 0008-5472
| pmid = 15256455
}}</ref>. Inhibition of h-ras was found to increase cisplatin sensitivity in glioblastoma cells<ref>{{Cite journal
| last = Messina
| first = Samantha
| last2 = Leonetti
| first2 = Carlo
| last3 = De Gregorio
| first3 = Giorgia
| last4 = Affatigato
| first4 = Valentina
| last5 = Ragona
| first5 = Giuseppe
| last6 = Frati
| first6 = Luigi
| last7 = Zupi
| first7 = Gabriella
| last8 = Santoni
| first8 = Angela
| last9 = Porcellini
| first9 = Antonio
| date = 2004-07-23
| title = Ras inhibition amplifies cisplatin sensitivity of human glioblastoma
| url = http://www.sciencedirect.com/science/article/pii/S0006291X04012306
| journal = Biochemical and Biophysical Research Communications
| volume = 320
| issue = 2
| pages = 493–500
| doi = 10.1016/j.bbrc.2004.06.003
}}</ref>. Upregulated expression ''[[Bcl-2]]'' in leukemic cells ([[Non-Hodgkin lymphoma|non-Hodgkin’s lymphoma]]) resulted in decreased levels of apoptosis in response to chemotherapeutic agents, as ''Bcl-2'' is a pro-survival [[oncogene]]<ref>{{Cite journal
| last = Miyashita
| first = T
| last2 = Reed
| first2 = JC
| date = January 1, 1993
| title = Bcl-2 oncoprotein blocks chemotherapy-induced apoptosis in a human leukemia cell line
| url = http://www.bloodjournal.org/content/81/1/151.short?sso-checked=true
| journal = Blood: 81 (1)
| doi =
| pmid = 8417786
| access-date =
}}</ref>.

=== Alteration of Target Molecules ===
During [[targeted therapy]], oftentimes the target of the therapy is modified and its expression decreased to the point that it is no longer an effective target for therapy. One example of this is the loss of [[estrogen receptor]] (ER) and [[progesterone receptor]] (PR) upon [[Antiestrogen|anti-estrogen treatment]] of breast cancer<ref name=":2">{{Cite journal
| last = Clarke
| first = Robert
| last2 = Liu
| first2 = Minetta C.
| last3 = Bouker
| first3 = Kerrie B.
| last4 = Gu
| first4 = Zhiping
| last5 = Lee
| first5 = Richard Y.
| last6 = Zhu
| first6 = Yuelin
| last7 = Skaar
| first7 = Todd C.
| last8 = Gomez
| first8 = Bianca
| last9 = O'Brien
| first9 = Kerry
| date = 2003-01-01
| title = Antiestrogen resistance in breast cancer and the role of estrogen receptor signaling
| url = http://www.nature.com/onc/journal/v22/n47/full/1206937a.html
| journal = Oncogene
| language = en
| volume = 22
| issue = 47
| pages = 7316–7339
| doi = 10.1038/sj.onc.1206937
| issn = 0950-9232
}}</ref>. Patients with loss of ER and PR are no longer receptive to [[tamoxifen]] or other anti-estrogen treatments, and while cancer cells remain somewhat responsive to [[estrogen]] synthesis inhibitors, they eventually become unresponsive to [[Endocrine system|endocrine]] manipulation and no longer dependent on estrogen for growth<ref name=":2" />.

Another line of therapeutics used for treating breast cancer is targeting of [[Kinase|kinases]] like [[HER2/neu|human epidermal growth factor receptor 2]] (HER2) from the [[Epidermal growth factor receptor|EGFR]] family. Mutations often occur in the ''HER2'' gene upon treatment with an inhibitor, with about 50% of patients with lung cancer found to have an ''EGFR-T790M'' gatekeeper mutation<ref name=":1" />.

Treatment of [[Chronic myelogenous leukemia|chronic myeloid leukemia]] (CML) involves a [[Tyrosine-kinase inhibitor|tyrosine kinase inhibitor]] that targets the ''[[Philadelphia chromosome|BCR/ABL]]'' fusion gene called [[Imatinib]]. Studies have found that in some patients that have become resistant to Imatinib, the ''BCR/ABL'' gene is reactivated or amplified, or a single [[point mutation]] has occurred on the gene. These point mutations enhance [[autophosphorylation]] of the BCR-ABL protein, resulting in the stabilization of the ATP-binding site into its active form, which cannot be bound by Imatinib for proper drug activation<ref>{{Cite journal
| last = Nardi
| first = Valentina
| last2 = Azam
| first2 = Mohammad
| last3 = Daley
| first3 = George Q.
| date = 2004-01-01
| title = Mechanisms and implications of imatinib resistance mutations in BCR-ABL
| url = http://www.ncbi.nlm.nih.gov/pubmed/14676625
| journal = Current Opinion in Hematology
| volume = 11
| issue = 1
| pages = 35–43
| issn = 1065-6251
| pmid = 14676625
}}</ref>.

[[Topoisomerase]] is a lucrative target for cancer therapy due to its critical role as an enzyme in [[DNA replication]], and many [[Topoisomerase inhibitor|topoisomerase inhibitors]] have been made<ref name=":3">{{Cite journal
| last = Ganapathi
| first = Ram
| last2 = Ganapathi
| first2 = Mahrukh K.
| date = 2013-01-01
| title = Mechanisms regulating resistance to inhibitors of topoisomerase II
| url = http://journal.frontiersin.org/article/10.3389/fphar.2013.00089/abstract
| journal = Pharmacology of Anti-Cancer Drugs
| volume = 4
| pages = 89
| doi = 10.3389/fphar.2013.00089
| pmc = 3729981
| pmid = 23914174
}}</ref>. Resistance can occur when topoisomerase levels are decreased, or when different isoforms of topoisomerase are differentially distributed within the cell<ref name=":3" />. Mutant enzymes have also been reported in patient leukemic cells, as well as mutations in other cancers that confer resistance to topoisomerase inhibitors<ref name=":3" />.

=== Protein and Pathway Mechanisms ===
One of the mechanisms of antineoplastic resistance is over-expression of drug-metabolizing enzymes or carrier molecules<ref name=":0" />. By increasing expression of metabolic enzymes, drugs are more rapidly converted to drug conjugates or inactive forms that can then be excreted. For example, increased expression of [[glutathione]] promotes drug resistance, as the electrophilic properties of glutathione allow it to react with cytotoxic agents, inactivating them<ref>{{Cite journal
| last = Schröder
| first = Carolina P.
| last2 = Godwin
| first2 = Andrew K.
| last3 = O'dwyer
| first3 = Peter J.
| last4 = Tew
| first4 = Kenneth D.
| last5 = Hamilton
| first5 = Thomas C.
| last6 = Ozols
| first6 = Robert F.
| date = 1996-01-01
| title = Glutathione and Drug Resistance
| url = http://dx.doi.org/10.3109/07357909609018891
| journal = Cancer Investigation
| volume = 14
| issue = 2
| pages = 158–168
| doi = 10.3109/07357909609018891
| issn = 0735-7907
| pmid = 8597901
}}</ref>. In some cases, decreased expression or loss of expression of drug-metabolising enzymes confers resistance, as the enzymes are needed to process a drug from an inactive form to an active form. [[Arabinose|Arabinoside]], a commonly used chemotherapy for leukemia and lymphomas, is converted into cytosine arabinoside triphosphate by deoxycytidine kinase. Mutation of deoxycytidine kinase or loss of expression results in resistance to arabinoside<ref name=":0" />. This is a form of enzymatic deactivation.

Growth factor expression levels can also promote resistance to antineoplastic therapies<ref name=":0" />. In breast cancer, drug resistant cells were found to express high levels of IL-6, while sensitive cells did not express significant levels of the growth factor. IL-6 activates the CCAAT enhancer-binding protein transcription factors which activate ''MDR1'' gene expression (see Alteration of Membrane Transport)<ref>{{Cite journal
| last = Conze
| first = D.
| last2 = Weiss
| first2 = L.
| last3 = Regen
| first3 = P. S.
| last4 = Bhushan
| first4 = A.
| last5 = Weaver
| first5 = D.
| last6 = Johnson
| first6 = P.
| last7 = Rincón
| first7 = M.
| date = 2001-12-15
| title = Autocrine production of interleukin 6 causes multidrug resistance in breast cancer cells
| url = http://www.ncbi.nlm.nih.gov/pubmed/11751408
| journal = Cancer Research
| volume = 61
| issue = 24
| pages = 8851–8858
| issn = 0008-5472
| pmid = 11751408
}}</ref>.

== Genomic Mechanisms underlying Acquired Resistance ==

=== Genome Alterations ===
[[Chromosomal rearrangement]] due to genome instability can cause gene amplification and deletion, both of which underlies the development of multidrug resistance.
* [[Gene duplication|Gene amplification]] is the increase in copy number of a region of a chromosome<ref name=":4">{{Cite journal
| last = Albertson
| first = Donna G.
| date = 2006-08-01
| title = Gene amplification in cancer
| url = http://www.cell.com/article/S0168952506001752/abstract
| journal = Trends in Genetics
| language = English
| volume = 22
| issue = 8
| pages = 447–455
| doi = 10.1016/j.tig.2006.06.007
| issn = 0168-9525
| pmid = 16787682
}}</ref>. Such event occurs frequently in solid tumors, and can contribute to tumor evolution through altered gene expression<ref name=":4" />. The initiation of gene amplification is on the basis of genome instability. Research on Chinese hamster cells showed that amplifications in [[Dihydrofolate reductase|DHFR]] gene (involved in DNA synthesis) began with chromosome break in distal areas to the gene, and subsequent cycles of [[Breakage-fusion-bridge cycle|bridge-breakage-fusion]] formations result in large intrachromosomal repeats<ref>{{Cite journal
| last = Ma
| first = C.
| last2 = Martin
| first2 = S.
| last3 = Trask
| first3 = B.
| last4 = Hamlin
| first4 = J. L.
| date = 1993-04-01
| title = Sister chromatid fusion initiates amplification of the dihydrofolate reductase gene in Chinese hamster cells.
| url = http://genesdev.cshlp.org/content/7/4/605
| journal = Genes & Development
| language = en
| volume = 7
| issue = 4
| pages = 605–620
| doi = 10.1101/gad.7.4.605
| issn = 0890-9369
| pmid = 8458577
}}</ref>. The over amplification of oncogenes can occur in response to drug administration, and this is thought to be the underlying mechanism in several class of resistance<ref name=":4" />. For example, ''DHFR'' amplification occurs in response to [[methotrexate]]<ref>Gorlick, R. et al. (1996) Intrinsic and acquired resistance to methotrexate in acute leukemia. New Engl. J. Med. 335, 1041–1048</ref>, [[Thymidylate synthase|''TYMS'']] (involved in DNA synthesis) amplification occurs in response to [[Fluorouracil|5-fluorouracil]]<ref>{{Cite journal
| last = Wang
| first = Tian-Li
| last2 = Diaz
| first2 = Luis A.
| last3 = Romans
| first3 = Katharine
| last4 = Bardelli
| first4 = Alberto
| last5 = Saha
| first5 = Saurabh
| last6 = Galizia
| first6 = Gennaro
| last7 = Choti
| first7 = Michael
| last8 = Donehower
| first8 = Ross
| last9 = Parmigiani
| first9 = Giovanni
| date = 2004-03-02
| title = Digital karyotyping identifies thymidylate synthase amplification as a mechanism of resistance to 5-fluorouracil in metastatic colorectal cancer patients
| url = http://www.ncbi.nlm.nih.gov/pubmed/14970324
| journal = Proceedings of the National Academy of Sciences of the United States of America
| volume = 101
| issue = 9
| pages = 3089–3094
| doi = 10.1073/pnas.0308716101
| issn = 0027-8424
| pmc = 420348
| pmid = 14970324
}}</ref>, and ''BCR-ABL'' amplification occurs in response to [[Imatinib|Imatinib mesylate]]<ref>{{Cite journal
| last = Gorre
| first = M. E.
| last2 = Mohammed
| first2 = M.
| last3 = Ellwood
| first3 = K.
| last4 = Hsu
| first4 = N.
| last5 = Paquette
| first5 = R.
| last6 = Rao
| first6 = P. N.
| last7 = Sawyers
| first7 = C. L.
| date = 2001-08-03
| title = Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification
| url = http://www.ncbi.nlm.nih.gov/pubmed/11423618
| journal = Science (New York, N.Y.)
| volume = 293
| issue = 5531
| pages = 876–880
| doi = 10.1126/science.1062538
| issn = 0036-8075
| pmid = 11423618
}}</ref>. Determining areas of gene amplification using techniques such as FISH ([[fluorescence in situ hybridization]]) has huge clinical implications.
* [[Deletion (genetics)|Gene deletion]] is the opposite of gene amplification. It is where a region of a chromosome is lost. Gene deletion confers drug resistance through the loss of tumor suppressor genes such as TP53<ref name=":0" />.

=== Epigenetic Mechanisms ===
The involvement of [[Epigenetics|epigenetic]] modifications in antineoplastic drug resistance have not been discussed to the same extent as those above mechanisms. Nonetheless epigenetic modifications plays a major role in cancer development and drug resistance as they regulate gene expression<ref name=":1" />. Two main types of epigenetic controls are DNA methylation and histone methylation/acetylation. [[DNA methylation]] is the process of adding methyl groups to DNA, usually in the upstream [[Promoter (genetics)|promoter]] regions, which stops DNA transcription at the region and effectively silences individual genes. [[Histone|Histone modifications]], such as [[deacetylation]], alters [[chromatin]] formation and silence large chromosomal regions. In cancer cells, where normal regulation of gene expression breaks down, the oncogenes are activated via hypomethylation and tumor suppressors are silenced via hypermethylation. Similarly in drug resistance development, it has been suggested that epigenetic modifications can result in the activation and overexpression of pro drug resistance genes.<ref name=":1" />

Studies on cancer cell lines have shown that hypomethylation (loss of methylation) of the ''MDR-1'' gene promoter resulted in its overexpression and the acquisition of multidrug resistance<ref>Kantharidis, P.; El-Oska, A.; de Silva, M.; Wall, D.M.; Hu, X.F.; Slater, A.; Nadalin, G.; Parkin, J.D.; Zalcberg, J.R. Altered methylation of the human MDR1 promoter is associated with acquired multidrug resistance. Clin. Cancer Res. 1997, 3, 2025–2032.</ref>.

In a methotrexate resistant breast cancer cell line lacking drug uptake and folate carrier expression, administration of [[Decitabine|DAC]], a DNA methylation inhibitor, effectively improved drug uptake and folate carrier expression<ref>{{Cite journal
| last = Worm
| first = J.
| last2 = Kirkin
| first2 = A. F.
| last3 = Dzhandzhugazyan
| first3 = K. N.
| last4 = Guldberg
| first4 = P.
| date = 2001-10-26
| title = Methylation-dependent silencing of the reduced folate carrier gene in inherently methotrexate-resistant human breast cancer cells
| url = http://www.ncbi.nlm.nih.gov/pubmed/11509559
| journal = The Journal of Biological Chemistry
| volume = 276
| issue = 43
| pages = 39990–40000
| doi = 10.1074/jbc.M103181200
| issn = 0021-9258
| pmid = 11509559
}}</ref>.

Acquired resistance to the alkylating drug [[fotemustine]] in [[melanoma]] cell showed high MGMT activity related to the [[hypermethylation]] of the ''MGMT'' gene exons<ref>{{Cite journal
| last = Christmann
| first = Markus
| last2 = Pick
| first2 = Matthias
| last3 = Lage
| first3 = Hermann
| last4 = Schadendorf
| first4 = Dirk
| last5 = Bernd Kaina
| date = 2001-04-01
| title = Acquired resistance of melanoma cells to the antineoplastic agent fotemustine is caused by reactivation of the DNA repair gene mgmt
| url = http://onlinelibrary.wiley.com/doi/10.1002/1097-0215(200102)9999:99993.0.CO;2-V/abstract
| journal = International Journal of Cancer
| language = en
| volume = 92
| issue = 1
| pages = 123–129
| doi = 10.1002/1097-0215(200102)9999:99993.0.CO;2-V
| issn = 1097-0215
}}</ref>.

In Imatinib resistant cell lines, silencing of the ''[[SOCS3|SOCS-3]]'' gene via methylation has been shown to cause [[STAT3]] protein activation, which resulted in uncontrolled proliferation<ref>{{Cite journal
| last = Al-Jamal
| first = Hamid A. N.
| last2 = Jusoh
| first2 = Siti Asmaa Mat
| last3 = Yong
| first3 = Ang Cheng
| last4 = Asan
| first4 = Jamaruddin Mat
| last5 = Hassan
| first5 = Rosline
| last6 = Johan
| first6 = Muhammad Farid
| date = 2014-01-01
| title = Silencing of suppressor of cytokine signaling-3 due to methylation results in phosphorylation of STAT3 in imatinib resistant BCR-ABL positive chronic myeloid leukemia cells
| url = http://www.ncbi.nlm.nih.gov/pubmed/24969884
| journal = Asian Pacific journal of cancer prevention: APJCP
| volume = 15
| issue = 11
| pages = 4555–4561
| issn = 1513-7368
| pmid = 24969884
}}</ref>.

== Genetic Approaches to Overcome Drug Resistance ==
MDR proteins are known to be drug-resistance genes, and are highly expressed in various cancers. Inhibition of the ''MDR'' genes could result in sensitization of cells to therapeutics and a decrease in antineoplastic resistance. Reversin 121 (R121) is a high-affinity peptide for MDR, and  use of R121 as a treatment for pancreatic cancer cells results in increased chemosensitivity and decreased proliferation<ref>{{Cite journal
| last = Hoffmann
| first = Katrin
| last2 = Bekeredjian
| first2 = Raffi
| last3 = Schmidt
| first3 = Jan
| last4 = B&uuml;chler
| first4 = Markus W.
| last5 = M&auml;rten
| first5 = Angela
| title = Effects of the High-Affinity Peptide Reversin 121 on Multidrug Resistance Proteins in Experimental Pancreatic Cancer
| url = http://www.karger.com/doi/10.1159/000178142
| journal = Tumor Biology
| volume = 29
| issue = 6
| pages = 351–358
| doi = 10.1159/000178142
}}</ref>.

Aberrant NF-κB expression is found in many cancers, and NF-κB has been found to be involved in resistance to platinum-based chemotherapies, such as cisplatin. NF-κB inhibition by genistein in various cancer cell lines (prostate, breast, lung and pancreas) showed increased growth inhibition and an increase in chemosensitivity, seen as an increase in apoptosis induced by therapeutic agents<ref>{{Cite journal
| last = Li
| first = Yiwei
| last2 = Ahmed
| first2 = Fakhara
| last3 = Ali
| first3 = Shadan
| last4 = Philip
| first4 = Philip A.
| last5 = Kucuk
| first5 = Omer
| last6 = Sarkar
| first6 = Fazlul H.
| date = 2005-08-01
| title = Inactivation of Nuclear Factor κB by Soy Isoflavone Genistein Contributes to Increased Apoptosis Induced by Chemotherapeutic Agents in Human Cancer Cells
| url = http://cancerres.aacrjournals.org/content/65/15/6934
| journal = Cancer Research
| language = en
| volume = 65
| issue = 15
| pages = 6934–6942
| doi = 10.1158/0008-5472.CAN-04-4604
| issn = 0008-5472
| pmid = 16061678
}}</ref>. However, targeting the NF-κB pathway can be difficult, as there could be many off-target and non-specific effects.

Expression of ''TP53'' causes defects in the apoptotic pathway, allowing cancerous cells to avoid death. Re-expression of the wild-type gene in cancer cells has been shown to inhibit cell proliferation, induce cell cycle arrest and apoptosis<ref>{{Cite journal
| last = Liu
| first = Xiangrui
| last2 = Wilcken
| first2 = Rainer
| last3 = Joerger
| first3 = Andreas C.
| last4 = Chuckowree
| first4 = Irina S.
| last5 = Amin
| first5 = Jahangir
| last6 = Spencer
| first6 = John
| last7 = Fersht
| first7 = Alan R.
| date = 2013-07-01
| title = Small molecule induced reactivation of mutant p53 in cancer cells
| url = http://nar.oxfordjournals.org/content/41/12/6034
| journal = Nucleic Acids Research
| language = en
| volume = 41
| issue = 12
| pages = 6034–6044
| doi = 10.1093/nar/gkt305
| issn = 0305-1048
| pmc = 3695503
| pmid = 23630318
}}</ref>.

In [[ovarian cancer]], the ''ATP7B'' gene encodes for a copper efflux transporter, found to be upregulated in cisplatin-resistant cell lines and tumors. Development of antisense deoxynucleotides against ATP7B mRNA and treatment of an ovarian cancer cell line shows that inhibition of ATP7B increases sensitivity of the cells to cisplatin<ref>{{Cite journal
| last = Xu
| first = W.
| last2 = Cai
| first2 = B.
| last3 = Chen
| first3 = J.l.
| last4 = Li
| first4 = L.x.
| last5 = Zhang
| first5 = J.r.
| last6 = Sun
| first6 = Y.y.
| last7 = Wan
| first7 = X.p.
| date = 2008-07-01
| title = ATP7B antisense oligodeoxynucleotides increase the cisplatin sensitivity of human ovarian cancer cell line SKOV3ipl
| url = http://onlinelibrary.wiley.com/doi/10.1111/j.1525-1438.2007.01085.x/abstract
| journal = International Journal of Gynecological Cancer
| language = en
| volume = 18
| issue = 4
| pages = 718–722
| doi = 10.1111/j.1525-1438.2007.01085.x
| issn = 1525-1438
}}</ref>.

== References ==
<references />


[[Category:Oncology]]
[[Category:Oncology]]

Revision as of 11:22, 25 February 2016

Antineoplastic resistance, often used interchangeably with chemotherapy resistance, is the multiple drug resistance of neoplastic (cancerous) cells, or the ability of cancer cells to survive and grow despite anti-cancer therapies.

There are two general causes of antineoplastic therapy failure[1]:

  1. Possession of inherent properties, such as genetic characteristics, which gave cancer cells their resistance[1]. Such idea is rooted in the concept of cancer cell heterogeneity.
  2. Acquired resistance after drug exposure[1]. Cancer cells have the ability to become resistant to multiple drugs by various mechanisms[1] [2]
    • Altered membrane transport
    • Enhanced DNA repair
    • Apoptotic pathway defects
    • Alteration of target molecules
    • Protein and pathway mechanisms such as enzymatic deactivation

As cancer is very much a disease of genetic mutations, underlying the development of acquired drug resistance mechanisms are several genomic events.

  • Genome alterations (e.g. gene amplification and deletion)
  • Epigenetic modifications

The mechanisms of antineoplastic resistance is in no way exhaustive. Cancer cells ultimately employ a variety of tools, involving genes, proteins and altered pathways, to ensure their survival against drugs. As such, the treatment of cancer is a very complex and challenging field.

Cancer Cell Heterogeneity

Cancer cell heterogeneity, or tumour heterogeneity is the idea that, within a tumor mass, a certain population of cancer cells exists that possess inherent genetic characteristics that confer drug resistance. The administration of antineoplastic drugs acts as a selection mechanism that kills non-resistant cells, while favoring these resistant cells. While the initial tumor mass may shrink as an initial response to the drug, these resistant colonies will survive treatment and then propagate, eventually causing a cancer relapse.

A slightly different way in which cancer cell heterogeneity gives rise to disease progression is via targeted therapies, which target a specific molecular marker. Tumor cells that do not express the specific marker are not killed, and are then able to divide and mutate further, creating a new heterogeneous tumor.

Nonetheless, the presence of such genomic heterogeneity complicates antineoplastic treatment.

An overview of antineoplastic resistance mechanisms

Mechanisms of Acquired Resistance

Alteration of Membrane Transport

Many classes of antineoplastic drugs acts on DNA, nuclear components, and intracellular pathways, meaning that they require entry into the cancer cells. The p-glycoprotein (P-gp), or the multiple drug resistance protein, is a phosphorylated and glycosylated membrane transporter that can shuttle drugs out of the cell, thereby decreasing or ablating drug efficacy. This transporter protein is encoded by the MDR1 gene and is also called the ATP-binding cassette (ABC) protein.  MDR1 has promiscuous substrate specificity, allowing it to be able to transport many structurally diverse compounds across the cell membrane, mainly hydrophobic compounds. Studies have found that the MDR1 gene can be activated and overexpressed in response to drug administration, thus forming the basis for resistance to many drugs[1]. Overexpression of the MDR1 gene in cancer cells is used to keep intracellular levels of antineoplastic drugs below cell-killing levels.

For example, rifampicin has been found to induce MDR1 expression. Experiments in differe

nt drug resistant cell lines and patient DNA revealed gene rearrangements that had initiated the activation or overexpression of MDR1[3]. A C3435T polymorphism in exon 226 of MDR1 has also been strongly correlated with p-glycoprotein activities[4].

Studies have shown that MDR1 activation occurs through activation of NF-κB, a protein complex that acts as a transcription factor[5][6]. There is a NF-κB binding site located adjacent to the rat mdr1b gene[7], and studies show that MDR1 upregulation is reliant on NF-κB activation as well[8][9]. NF-κB is active in tumour cells due to mutations in the NF-κB gene or in the inhibitory IκB gene. In colon cancer cells, inhibition of NF-κB or MDR1 resulted in increased apoptosis in response to a chemotherapeutic agent used to treat various cancers[10].

Enhanced DNA Repair

Enhanced DNA repair plays an important role in the ability for cancer cells to overcome drug-induced DNA damages.

Platinum-base chemotherapy, such as cisplatin, targets tumor cells by cross-linking their DNA strands, causing mutation and damage[1]. Such damage will trigger programmed cell death (apoptosis) in cancer cells. Cisplatin resistance occurs when cancer cells develop an enhanced ability to reverse such damage, by removing the cisplatin from DNA and repairing any damage done[1][2]. The unregulated expression of the excision repair cross-complementing (ERCC1) gene and protein in cisplatin-resistant tumors underlies such process[1].

Some chemotherapies are alkylating agents that result in the attachment of an alkyl group to DNA. O6-methylguanine DNA methyltransferase (MGMT) is a DNA repair enzyme that removes alkyl groups from DNA. Expression of MGMT is upregulated in many cancer cells, providing a protective function from alkylating agents[2]. Increased MGMT expression has been found in colon cancer, lung cancer, non-Hodgkin’s lymphoma, breast cancer, gliomas, myeloma and pancreatic cancer[11].

Apoptotic Pathway Defects

TP53 is a tumor suppressor gene that encodes the p53 protein. It is a protein that responds to DNA damage, either by initiating DNA repair, cell cycle arrest, or apoptosis. The loss of TP53 via gene deletion can result in continued replication despite DNA damage. This tolerance of DNA damage can grant cancer cells a method of resistance to drugs aiming to induce apoptosis via DNA damage[1][2].

Other genes involved in the apoptotic pathway related drug resistance includes h-ras and bcl-2/bax[12]. Oncogenic h-ras has been found to increase expression of ERCC1, resulting in enhanced DNA repair (see above)[13]. Inhibition of h-ras was found to increase cisplatin sensitivity in glioblastoma cells[14]. Upregulated expression Bcl-2 in leukemic cells (non-Hodgkin’s lymphoma) resulted in decreased levels of apoptosis in response to chemotherapeutic agents, as Bcl-2 is a pro-survival oncogene[15].

Alteration of Target Molecules

During targeted therapy, oftentimes the target of the therapy is modified and its expression decreased to the point that it is no longer an effective target for therapy. One example of this is the loss of estrogen receptor (ER) and progesterone receptor (PR) upon anti-estrogen treatment of breast cancer[16]. Patients with loss of ER and PR are no longer receptive to tamoxifen or other anti-estrogen treatments, and while cancer cells remain somewhat responsive to estrogen synthesis inhibitors, they eventually become unresponsive to endocrine manipulation and no longer dependent on estrogen for growth[16].

Another line of therapeutics used for treating breast cancer is targeting of kinases like human epidermal growth factor receptor 2 (HER2) from the EGFR family. Mutations often occur in the HER2 gene upon treatment with an inhibitor, with about 50% of patients with lung cancer found to have an EGFR-T790M gatekeeper mutation[2].

Treatment of chronic myeloid leukemia (CML) involves a tyrosine kinase inhibitor that targets the BCR/ABL fusion gene called Imatinib. Studies have found that in some patients that have become resistant to Imatinib, the BCR/ABL gene is reactivated or amplified, or a single point mutation has occurred on the gene. These point mutations enhance autophosphorylation of the BCR-ABL protein, resulting in the stabilization of the ATP-binding site into its active form, which cannot be bound by Imatinib for proper drug activation[17].

Topoisomerase is a lucrative target for cancer therapy due to its critical role as an enzyme in DNA replication, and many topoisomerase inhibitors have been made[18]. Resistance can occur when topoisomerase levels are decreased, or when different isoforms of topoisomerase are differentially distributed within the cell[18]. Mutant enzymes have also been reported in patient leukemic cells, as well as mutations in other cancers that confer resistance to topoisomerase inhibitors[18].

Protein and Pathway Mechanisms

One of the mechanisms of antineoplastic resistance is over-expression of drug-metabolizing enzymes or carrier molecules[1]. By increasing expression of metabolic enzymes, drugs are more rapidly converted to drug conjugates or inactive forms that can then be excreted. For example, increased expression of glutathione promotes drug resistance, as the electrophilic properties of glutathione allow it to react with cytotoxic agents, inactivating them[19]. In some cases, decreased expression or loss of expression of drug-metabolising enzymes confers resistance, as the enzymes are needed to process a drug from an inactive form to an active form. Arabinoside, a commonly used chemotherapy for leukemia and lymphomas, is converted into cytosine arabinoside triphosphate by deoxycytidine kinase. Mutation of deoxycytidine kinase or loss of expression results in resistance to arabinoside[1]. This is a form of enzymatic deactivation.

Growth factor expression levels can also promote resistance to antineoplastic therapies[1]. In breast cancer, drug resistant cells were found to express high levels of IL-6, while sensitive cells did not express significant levels of the growth factor. IL-6 activates the CCAAT enhancer-binding protein transcription factors which activate MDR1 gene expression (see Alteration of Membrane Transport)[20].

Genomic Mechanisms underlying Acquired Resistance

Genome Alterations

Chromosomal rearrangement due to genome instability can cause gene amplification and deletion, both of which underlies the development of multidrug resistance.

  • Gene amplification is the increase in copy number of a region of a chromosome[21]. Such event occurs frequently in solid tumors, and can contribute to tumor evolution through altered gene expression[21]. The initiation of gene amplification is on the basis of genome instability. Research on Chinese hamster cells showed that amplifications in DHFR gene (involved in DNA synthesis) began with chromosome break in distal areas to the gene, and subsequent cycles of bridge-breakage-fusion formations result in large intrachromosomal repeats[22]. The over amplification of oncogenes can occur in response to drug administration, and this is thought to be the underlying mechanism in several class of resistance[21]. For example, DHFR amplification occurs in response to methotrexate[23], TYMS (involved in DNA synthesis) amplification occurs in response to 5-fluorouracil[24], and BCR-ABL amplification occurs in response to Imatinib mesylate[25]. Determining areas of gene amplification using techniques such as FISH (fluorescence in situ hybridization) has huge clinical implications.
  • Gene deletion is the opposite of gene amplification. It is where a region of a chromosome is lost. Gene deletion confers drug resistance through the loss of tumor suppressor genes such as TP53[1].

Epigenetic Mechanisms

The involvement of epigenetic modifications in antineoplastic drug resistance have not been discussed to the same extent as those above mechanisms. Nonetheless epigenetic modifications plays a major role in cancer development and drug resistance as they regulate gene expression[2]. Two main types of epigenetic controls are DNA methylation and histone methylation/acetylation. DNA methylation is the process of adding methyl groups to DNA, usually in the upstream promoter regions, which stops DNA transcription at the region and effectively silences individual genes. Histone modifications, such as deacetylation, alters chromatin formation and silence large chromosomal regions. In cancer cells, where normal regulation of gene expression breaks down, the oncogenes are activated via hypomethylation and tumor suppressors are silenced via hypermethylation. Similarly in drug resistance development, it has been suggested that epigenetic modifications can result in the activation and overexpression of pro drug resistance genes.[2]

Studies on cancer cell lines have shown that hypomethylation (loss of methylation) of the MDR-1 gene promoter resulted in its overexpression and the acquisition of multidrug resistance[26].

In a methotrexate resistant breast cancer cell line lacking drug uptake and folate carrier expression, administration of DAC, a DNA methylation inhibitor, effectively improved drug uptake and folate carrier expression[27].

Acquired resistance to the alkylating drug fotemustine in melanoma cell showed high MGMT activity related to the hypermethylation of the MGMT gene exons[28].

In Imatinib resistant cell lines, silencing of the SOCS-3 gene via methylation has been shown to cause STAT3 protein activation, which resulted in uncontrolled proliferation[29].

Genetic Approaches to Overcome Drug Resistance

MDR proteins are known to be drug-resistance genes, and are highly expressed in various cancers. Inhibition of the MDR genes could result in sensitization of cells to therapeutics and a decrease in antineoplastic resistance. Reversin 121 (R121) is a high-affinity peptide for MDR, and  use of R121 as a treatment for pancreatic cancer cells results in increased chemosensitivity and decreased proliferation[30].

Aberrant NF-κB expression is found in many cancers, and NF-κB has been found to be involved in resistance to platinum-based chemotherapies, such as cisplatin. NF-κB inhibition by genistein in various cancer cell lines (prostate, breast, lung and pancreas) showed increased growth inhibition and an increase in chemosensitivity, seen as an increase in apoptosis induced by therapeutic agents[31]. However, targeting the NF-κB pathway can be difficult, as there could be many off-target and non-specific effects.

Expression of TP53 causes defects in the apoptotic pathway, allowing cancerous cells to avoid death. Re-expression of the wild-type gene in cancer cells has been shown to inhibit cell proliferation, induce cell cycle arrest and apoptosis[32].

In ovarian cancer, the ATP7B gene encodes for a copper efflux transporter, found to be upregulated in cisplatin-resistant cell lines and tumors. Development of antisense deoxynucleotides against ATP7B mRNA and treatment of an ovarian cancer cell line shows that inhibition of ATP7B increases sensitivity of the cells to cisplatin[33].

References

  1. ^ a b c d e f g h i j k l m Luqmani, Y.A. "Mechanisms of Drug Resistance in Cancer Chemotherapy". Medical Principles and Practice. 14 (1): 35–48. doi:10.1159/000086183.
  2. ^ a b c d e f g Housman, Genevieve; Byler, Shannon; Heerboth, Sarah; Lapinska, Karolina; Longacre, Mckenna; Snyder, Nicole; Sarkar, Sibaji (2014-09-05). "Drug Resistance in Cancer: An Overview". Cancers. 6 (3): 1769–1792. doi:10.3390/cancers6031769. PMC 4190567. PMID 25198391.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  3. ^ Mickley, L A; Spengler, B A; Knutsen, T A; Biedler, J L; Fojo, T (1997-04-15). "Gene rearrangement: a novel mechanism for MDR-1 gene activation". Journal of Clinical Investigation. 99 (8): 1947–1957. doi:10.1172/jci119362.
  4. ^ Hoffmeyer, S.; Burk, O.; Richter, O. von; Arnold, H. P.; Brockmöller, J.; Johne, A.; Cascorbi, I.; Gerloff, T.; Roots, I. (2000-03-28). "Functional polymorphisms of the human multidrug-resistance gene: Multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo". Proceedings of the National Academy of Sciences. 97 (7): 3473–3478. doi:10.1073/pnas.97.7.3473. ISSN 0027-8424.
  5. ^ Bentires-Alj, Mohamed; Barbu, Veronique; Fillet, Marianne; Chariot, Alain; Relic, Biserka; Jacobs, Nathalie; Gielen, Jacques; Merville, Marie-Paule; Bours, Vincent (2003-01-01). "NF-κB transcription factor induces drug resistance through MDR1 expression in cancer cells". Oncogene. 22 (1): 90–97. doi:10.1038/sj.onc.1206056. ISSN 0950-9232.
  6. ^ Kim, Hyung Gyun; Hien, Tran Thi; Han, Eun Hee; Hwang, Yong Pil; Choi, Jae Ho; Kang, Keon Wook; Kwon, Kwang-il; Kim, Bong-Hee; Kim, Sang Kyum (2011-03-01). "Metformin inhibits P-glycoprotein expression via the NF-κB pathway and CRE transcriptional activity through AMPK activation". British Journal of Pharmacology. 162 (5): 1096–1108. doi:10.1111/j.1476-5381.2010.01101.x. ISSN 1476-5381. PMC 3051382. PMID 21054339.
  7. ^ Zhou, Ge; Kuo, M. Tien (1997-06-13). "NF-κB-mediated Induction of mdr1b Expression by Insulin in Rat Hepatoma Cells". Journal of Biological Chemistry. 272 (24): 15174–15183. doi:10.1074/jbc.272.24.15174. ISSN 0021-9258.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  8. ^ Thévenod, Frank; Friedmann, Jenny M.; Katsen, Alice D.; Hauser, Ingeborg A. (2000-01-21). "Up-regulation of Multidrug Resistance P-glycoprotein via Nuclear Factor-κB Activation Protects Kidney Proximal Tubule Cells from Cadmium- and Reactive Oxygen Species-induced Apoptosis". Journal of Biological Chemistry. 275 (3): 1887–1896. doi:10.1074/jbc.275.3.1887. ISSN 0021-9258.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  9. ^ Kuo, Macus Tien; Liu, Zesheng; Wei, Yingjie; Lin-Lee, Yen-chiu; Tatebe, Shigeru; Mills, Gordon B.; Unate, Hitoshi (2002-03-27). "Induction of human MDR1 gene expression by 2-acetylaminofluorene is mediated by effectors of the phosphoinositide 3-kinase pathway that activate NF-kappaB signaling". Oncogene. 21 (13): 1945–1954. doi:10.1038/sj.onc.1205117. ISSN 0950-9232. PMID 11960367.
  10. ^ Bentires-Alj, Mohamed; Barbu, Veronique; Fillet, Marianne; Chariot, Alain; Relic, Biserka; Jacobs, Nathalie; Gielen, Jacques; Merville, Marie-Paule; Bours, Vincent (2003-01-01). "NF-κB transcription factor induces drug resistance through MDR1 expression in cancer cells". Oncogene. 22 (1): 90–97. doi:10.1038/sj.onc.1206056. ISSN 0950-9232.
  11. ^ Gerson, Stanton L. "MGMT: its role in cancer aetiology and cancer therapeutics". Nature Reviews Cancer. 4 (4): 296–307. doi:10.1038/nrc1319.
  12. ^ Burger, H.; Nooter, K.; Boersma, A. W.; van Wingerden, K. E.; Looijenga, L. H.; Jochemsen, A. G.; Stoter, G. (1999-05-17). "Distinct p53-independent apoptotic cell death signalling pathways in testicular germ cell tumour cell lines". International Journal of Cancer. Journal International Du Cancer. 81 (4): 620–628. ISSN 0020-7136. PMID 10225454.
  13. ^ Youn, Cha-Kyung; Kim, Mi-Hwa; Cho, Hyun-Ju; Kim, Hong-Beum; Chang, In-Youb; Chung, Myung-Hee; You, Ho Jin (2004-07-15). "Oncogenic H-Ras Up-Regulates Expression of ERCC1 to Protect Cells from Platinum-Based Anticancer Agents". Cancer Research. 64 (14): 4849–4857. doi:10.1158/0008-5472.CAN-04-0348. ISSN 0008-5472. PMID 15256455.
  14. ^ Messina, Samantha; Leonetti, Carlo; De Gregorio, Giorgia; Affatigato, Valentina; Ragona, Giuseppe; Frati, Luigi; Zupi, Gabriella; Santoni, Angela; Porcellini, Antonio (2004-07-23). "Ras inhibition amplifies cisplatin sensitivity of human glioblastoma". Biochemical and Biophysical Research Communications. 320 (2): 493–500. doi:10.1016/j.bbrc.2004.06.003.
  15. ^ Miyashita, T; Reed, JC (January 1, 1993). "Bcl-2 oncoprotein blocks chemotherapy-induced apoptosis in a human leukemia cell line". Blood: 81 (1). PMID 8417786.
  16. ^ a b Clarke, Robert; Liu, Minetta C.; Bouker, Kerrie B.; Gu, Zhiping; Lee, Richard Y.; Zhu, Yuelin; Skaar, Todd C.; Gomez, Bianca; O'Brien, Kerry (2003-01-01). "Antiestrogen resistance in breast cancer and the role of estrogen receptor signaling". Oncogene. 22 (47): 7316–7339. doi:10.1038/sj.onc.1206937. ISSN 0950-9232.
  17. ^ Nardi, Valentina; Azam, Mohammad; Daley, George Q. (2004-01-01). "Mechanisms and implications of imatinib resistance mutations in BCR-ABL". Current Opinion in Hematology. 11 (1): 35–43. ISSN 1065-6251. PMID 14676625.
  18. ^ a b c Ganapathi, Ram; Ganapathi, Mahrukh K. (2013-01-01). "Mechanisms regulating resistance to inhibitors of topoisomerase II". Pharmacology of Anti-Cancer Drugs. 4: 89. doi:10.3389/fphar.2013.00089. PMC 3729981. PMID 23914174.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  19. ^ Schröder, Carolina P.; Godwin, Andrew K.; O'dwyer, Peter J.; Tew, Kenneth D.; Hamilton, Thomas C.; Ozols, Robert F. (1996-01-01). "Glutathione and Drug Resistance". Cancer Investigation. 14 (2): 158–168. doi:10.3109/07357909609018891. ISSN 0735-7907. PMID 8597901.
  20. ^ Conze, D.; Weiss, L.; Regen, P. S.; Bhushan, A.; Weaver, D.; Johnson, P.; Rincón, M. (2001-12-15). "Autocrine production of interleukin 6 causes multidrug resistance in breast cancer cells". Cancer Research. 61 (24): 8851–8858. ISSN 0008-5472. PMID 11751408.
  21. ^ a b c Albertson, Donna G. (2006-08-01). "Gene amplification in cancer". Trends in Genetics. 22 (8): 447–455. doi:10.1016/j.tig.2006.06.007. ISSN 0168-9525. PMID 16787682.
  22. ^ Ma, C.; Martin, S.; Trask, B.; Hamlin, J. L. (1993-04-01). "Sister chromatid fusion initiates amplification of the dihydrofolate reductase gene in Chinese hamster cells". Genes & Development. 7 (4): 605–620. doi:10.1101/gad.7.4.605. ISSN 0890-9369. PMID 8458577.
  23. ^ Gorlick, R. et al. (1996) Intrinsic and acquired resistance to methotrexate in acute leukemia. New Engl. J. Med. 335, 1041–1048
  24. ^ Wang, Tian-Li; Diaz, Luis A.; Romans, Katharine; Bardelli, Alberto; Saha, Saurabh; Galizia, Gennaro; Choti, Michael; Donehower, Ross; Parmigiani, Giovanni (2004-03-02). "Digital karyotyping identifies thymidylate synthase amplification as a mechanism of resistance to 5-fluorouracil in metastatic colorectal cancer patients". Proceedings of the National Academy of Sciences of the United States of America. 101 (9): 3089–3094. doi:10.1073/pnas.0308716101. ISSN 0027-8424. PMC 420348. PMID 14970324.
  25. ^ Gorre, M. E.; Mohammed, M.; Ellwood, K.; Hsu, N.; Paquette, R.; Rao, P. N.; Sawyers, C. L. (2001-08-03). "Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification". Science (New York, N.Y.). 293 (5531): 876–880. doi:10.1126/science.1062538. ISSN 0036-8075. PMID 11423618.
  26. ^ Kantharidis, P.; El-Oska, A.; de Silva, M.; Wall, D.M.; Hu, X.F.; Slater, A.; Nadalin, G.; Parkin, J.D.; Zalcberg, J.R. Altered methylation of the human MDR1 promoter is associated with acquired multidrug resistance. Clin. Cancer Res. 1997, 3, 2025–2032.
  27. ^ Worm, J.; Kirkin, A. F.; Dzhandzhugazyan, K. N.; Guldberg, P. (2001-10-26). "Methylation-dependent silencing of the reduced folate carrier gene in inherently methotrexate-resistant human breast cancer cells". The Journal of Biological Chemistry. 276 (43): 39990–40000. doi:10.1074/jbc.M103181200. ISSN 0021-9258. PMID 11509559.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  28. ^ Christmann, Markus; Pick, Matthias; Lage, Hermann; Schadendorf, Dirk; Bernd Kaina (2001-04-01). "Acquired resistance of melanoma cells to the antineoplastic agent fotemustine is caused by reactivation of the DNA repair gene mgmt". International Journal of Cancer. 92 (1): 123–129. doi:10.1002/1097-0215(200102)9999:99993.0.CO;2-V. ISSN 1097-0215.
  29. ^ Al-Jamal, Hamid A. N.; Jusoh, Siti Asmaa Mat; Yong, Ang Cheng; Asan, Jamaruddin Mat; Hassan, Rosline; Johan, Muhammad Farid (2014-01-01). "Silencing of suppressor of cytokine signaling-3 due to methylation results in phosphorylation of STAT3 in imatinib resistant BCR-ABL positive chronic myeloid leukemia cells". Asian Pacific journal of cancer prevention: APJCP. 15 (11): 4555–4561. ISSN 1513-7368. PMID 24969884.
  30. ^ Hoffmann, Katrin; Bekeredjian, Raffi; Schmidt, Jan; Büchler, Markus W.; Märten, Angela. "Effects of the High-Affinity Peptide Reversin 121 on Multidrug Resistance Proteins in Experimental Pancreatic Cancer". Tumor Biology. 29 (6): 351–358. doi:10.1159/000178142.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  31. ^ Li, Yiwei; Ahmed, Fakhara; Ali, Shadan; Philip, Philip A.; Kucuk, Omer; Sarkar, Fazlul H. (2005-08-01). "Inactivation of Nuclear Factor κB by Soy Isoflavone Genistein Contributes to Increased Apoptosis Induced by Chemotherapeutic Agents in Human Cancer Cells". Cancer Research. 65 (15): 6934–6942. doi:10.1158/0008-5472.CAN-04-4604. ISSN 0008-5472. PMID 16061678.
  32. ^ Liu, Xiangrui; Wilcken, Rainer; Joerger, Andreas C.; Chuckowree, Irina S.; Amin, Jahangir; Spencer, John; Fersht, Alan R. (2013-07-01). "Small molecule induced reactivation of mutant p53 in cancer cells". Nucleic Acids Research. 41 (12): 6034–6044. doi:10.1093/nar/gkt305. ISSN 0305-1048. PMC 3695503. PMID 23630318.
  33. ^ Xu, W.; Cai, B.; Chen, J.l.; Li, L.x.; Zhang, J.r.; Sun, Y.y.; Wan, X.p. (2008-07-01). "ATP7B antisense oligodeoxynucleotides increase the cisplatin sensitivity of human ovarian cancer cell line SKOV3ipl". International Journal of Gynecological Cancer. 18 (4): 718–722. doi:10.1111/j.1525-1438.2007.01085.x. ISSN 1525-1438.