Survey
* Your assessment is very important for improving the work of artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the work of artificial intelligence, which forms the content of this project
1 Gene Therapy and Modification as a Therapeutic Strategy for Cancer 2 Abstract: 210 words 3 Text body: 3070 words 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 1 25 Abstract 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 Genetic therapy and modification is an exciting new paradigm of personalized medicine, allowing for medical procedures that can target diseases such as cancer in novel ways. Technologies which involve gene transfer treatments allow for the insertion of foreign DNA to affect or restore protein expression, and can alter the function of tumor cells. Gene therapy can also be used as a form of immunotherapy, either by modifying cancer cells to make them better targeted by the immune system, or by modifying the body’s immune cells to make them more aggressive towards tumours. Additionally, oncolytic virotherapy uses classes of genetically modified viruses that can specifically target and interfere with tumor cells. The ongoing development of the CRISPR-Cas9 gene editing tool may also have promise in future therapeutic applications, with the tool being capable of destroying cancer-causing latent viral infections like HPV from afflicted cells. Nonetheless, there are still many questions of safety, efficacy, and commercial viability which remain to be resolved with many gene therapy procedures, as well as the emerging controversy over the ethical, legal, and moral implications that modifying the genetic content of human beings will have on society. These concerns must also be confronted and addressed if the benefits promised by gene therapy are to be properly realized. 41 Introduction 42 Cancer is one of the most chronic and pressing health issues in the world today, causing 43 over 8 million deaths per year worldwide [1]. While existing surgical, radiation, and 44 chemotherapeutical procedures have improved outcomes in some cancer types, many cancers still 45 do not respond well to treatment [2]. Moreover, the overall prevalence and incidence rates of 46 cancer are expected to continue to rise [2,3]. Consequently, there is a great impetus to explore 47 novel approaches to treat the different cancers in ways that current standard approaches cannot. In 48 the past decade, the emerging field of gene therapy has arisen as a possible avenue to produce 49 innovative treatment options against cancer [4]. 50 Gene therapy focuses on using targeted delivery of genetic material and methods of genetic 51 modification to modify cells or tissues in order produce a positive therapeutic outcome [5]. The 52 advantage of gene therapy lies in its ability to enable new kinds of medical procedures and a wide 53 range of treatment options that were not previously possible. These include the insertion of foreign 54 DNA into target cells to affect or restore protein expression, targeting viruses to abnormal cells for 55 lysis and death, and even repairing deleterious genetic mutations [5]. Consequently, gene therapy 2 56 may prove to be an essential tool in personalized medicine, as it can design treatments that are 57 specific to a patient’s genetic composition. Although many of these techniques are largely 58 experimental and/or still in clinical trials, the progress in the field in the last decade has been quite 59 strong, with some gene therapy treatments starting to become commercially available. In the 60 context of cancer research, recent developments such as cancer vaccines, oncolytic virotherapy, 61 and gene transfer treatments are emerging as promising technologies to treat certain cancers with 62 a better efficacy and safety profile than current standard treatment modalities [5]. 63 This review aims to describe the current state of gene therapy research into applications for 64 cancer, to highlight the relative advantage and disadvantage they have over existing therapeutic 65 options, and to address the limitations currently being faced by the field and future directions 66 needed to overcome them. 67 Gene Transfer Treatments 68 Currently, one of the most widespread and well-established methods of gene therapy is the 69 insertion of foreign genes into target cells through a number of different transfer methods, 70 collectively called ‘gene transfer treatments’. One common approach is through viral vectors, 71 usually belonging to a class of viruses called adenoviruses, which carry and release the therapeutic 72 gene into a target tissue [5]. These adenoviruses are advantageous due to the ease in which foreign 73 genes can be inserted into their viral DNA, the relatively mild immune response it provokes from 74 the host, an exceedingly low rate of integration into the host’s chromosome that decreases the 75 possibility of unwanted mutations, and an inability to replicate (replication-incompetent) so that it 76 will not continue its lytic cycle spread to other cells [6]. 77 Indeed, many adenoviral vectors have been developed for use in the treatment of cancer, 78 and have generated a remarkable deal of excitement over their therapeutic impact. The drug 3 79 Gendicine, which became the first commercially approved gene therapy treatment in the world in 80 2003, is a recombinant adenovirus that contains the gene for the tumour suppressor p53 [7]. 81 Delivery of Gendicine will overexpress the p53 gene in tumour cells, or restore its activity in 82 tumour cells with a mutated and dysfunctional p53 gene [7]. Gendicine was a landmark in the 83 history of gene therapy for cancer, particularly squamous cell carcinoma, as it stimulated apoptosis 84 in tumours, increased the expression of other tumour suppressor genes, decreased the prevalence 85 of multi-drug resistance factors, and reduced vascular growth towards the cancerous tissues—all 86 while producing less side effects than conventional chemotherapy and radiotherapy [7]. There are 87 other types of viral classes that are available as well, such as retroviruses or adeno-associated 88 viruses, and each has their own strengths and effectiveness for different cell types/purposes. For 89 example, the drug Rexin-G is a specially designed retrovirus that integrates into the genome of 90 pancreatic tumour cells [8]. It carries a modified gene encoding a construct that interferes with 91 cyclin G1, thereby causing cell cycle arrest or death [8]. Rexin-G is showing promise for advanced 92 pancreatic cancer, including an increased mean survival time by almost 10 months compared to 93 standard treatment, and is currently in Phase III trials [8,9]. 94 However, there are many limitations and hurdles that remain with gene transfer treatments. 95 In 2003, attempts to treat the rare disorder X-linked severe combined immune-deficiency (SCID- 96 X1) using a retrovirus based agent unfortunately led to the development of T-cell leukemia in one 97 patient due to integration of the virus with the patient’s genome that caused genetic instability 98 leading to cancer [10]. This highlighted the need to develop vectors which took extraordinary 99 precautions to avoid integrating with host DNA in undesired regions, which is why most vectors 100 used today are adenoviruses or adeno-associated viruses. Nevertheless, the transition to these kind 101 of viruses has brought about its own unique share of problems. It has proven difficult to achieve 4 102 viral infection of a sufficient number of cells in target tissues to produce a clinically meaningful 103 response for many types of tumours. It has been observed that many viruses oftentimes end up 104 collecting in the liver for unknown reasons, thus reducing efficiency and causing a potentially 105 harmful immune response [5]. Further complicating this, since the viruses do not replicate, 106 additional viral loads must be injected periodically to sustain the expression of the therapeutic 107 gene. However, by the second time that the virus is introduced, the body has already developed an 108 acquired immunity to it and neutralizes much of the virus, reducing its efficiency even further [11]. 109 Currently, the delivery method is the most crucial factor underpinning the success of gene 110 transfer treatments. Yet, a remarkable degree of progress is being made in this area lately, with the 111 engineering of viral vectors becoming more sophisticated, as well as the emergence of non-viral 112 means of transfer, such as the insertion of naked DNA directly into cells [12,13]. Further advances 113 in this will significantly improve the ability to safely and precisely alter cellular function in order 114 to achieve the desired clinical outcome. 115 Immunotherapy 116 Gene therapy can also be used to modify a patient’s immune system in order to strengthen 117 the response against cancer cells. Those treatments which boost the immune system’s ability to 118 better target and destroy cancer cells are referred to as immunotherapy, and have been an aim of 119 cancer treatment for more than a century [14]. However, traditional immunotherapy based on 120 conventional technologies has proven to be limited, as cancer cells tend to evolve mechanisms 121 which help them evade immune detection. A number of different gene therapy techniques are being 122 explored as methods to overcome this limitation [5]. 123 A particularly interesting approach in this area is the development of genetically modified 124 ‘cancer vaccines’. Unlike conventional vaccines for infectious diseases which utilize agents that 125 help the body prevent the illness, cancer vaccines aim to cure or contain cancerous growth by 5 126 delivering material which trains the immune system to better recognize and attack present cancer 127 cells [4,15,16]. This material is created by harvesting cancer cells from the patient’s body, and 128 genetically modifying them through the addition of genes which produce antigenic and 129 immunostimulatory proteins; these are usually genes which encode for cytokines and pro- 130 inflammatory molecules [5]. The idea of this technique is essentially to produce modified cellular 131 debris from the cancer cells that contains more potent antigenic factors than the endogenous 132 tumour antigens. When this is delivered back into the patient, the co-stimulatory effects expressed 133 with the modified tumour antigens increases the activity of antigen-presenting cells and cytotoxic 134 T lymphocytes, thereby creating a stronger and more aggressive immune response against the 135 tumours [16]. Alternatively, a variation of this approach entails delivering the immunostimulatory 136 and antigenic protein genes directly to the cancer cells in the body, which increases immune 137 recognition of tumour cells and a more localized immune response [5]. 138 There are a number of genetically modified cancer vaccines being tested in clinical trials, 139 and many are showing a great deal of promise. A prominent example is GVAX, a vaccine that 140 targets advanced pancreatic cancer and consists of tumour cells modified to express granulocyte- 141 macrophage colony-stimulating factor (GM-SCF) [17,18]. The presence of GM-CSF on injected 142 cells containing tumour antigens stimulates the release of cytokines at the injection site which 143 activate antigen-presenting cells, CD4+, and CD8+ T cells to better recognize the circulating 144 tumor-associated antigens and strengthen the tumor-specific immune response [18]. The vaccine 145 is currently in Phase II trials, and recent studies have demonstrated a significant increase on patient 146 survival for cases of metastatic pancreatic adenocarcinoma as compared to standard 147 chemotherapeutic treatments [19]. Most of the side effects observed have been limited to minor 148 injection site reactions or flu-like symptoms [17,19]. Moreover, another advantage of the vaccine 6 149 is that it can be specifically designed for the patient using their tumour cells (called an ‘autologous’ 150 vaccine), thereby increasing its specificity for the patient’s unique immunological environment. 151 While the previously described concept focused on modifying the cancer cells to induce a 152 more potent immune response, another (and more unique) strategy is to modify the body’s immune 153 cells directly to make them more aggressive towards tumour cells [20]. This is done by extracting 154 and culturing lymphocytes from a patient’s body, and then genetically engineering them to 155 overexpress potent cytokines like Interleukin-2 (IL-2), or to produce T-cell receptors (TCRs) that 156 are specific for antigens on certain tumours [15,20]. A successful example of this is the use of T- 157 cells engineered to express TCRs against the NY-ESO-1 antigen in patients with metastatic 158 melanoma and metastatic synovial cell sarcoma [21]. Trials have shown that 50-80% of patients 159 demonstrate objectively better regression of the cancer compared to conventional chemotherapy, 160 with no reported toxic side effects against other tissues [21]. One caveat of this technology however 161 is that it is currently incredibly expensive; the average cost for a patient to have their T-cells 162 genetically engineered is about USD 75,000. 163 Overall, immunotherapy for cancer treatment is rapidly developing, and may be a source 164 of a large deal of breakthroughs against cancer in the near future. The main hurdles currently are 165 the immense cost and time needed to produce autologous vaccines and genetically engineered T- 166 cells. This is improving as more pharmaceutical companies are investing into the area and 167 developing increasingly efficient methods of production, as well as through the use of allogenic 168 cancer cells (derived from cultured cell lines) to reduce the amount of autologous cells needed [5]. 169 Oncolytic Virotherapy 170 A related concept to immunotherapy is the use of genetically engineered viral particles 171 which specifically recognize and attack cancerous tumours, called oncolytic virotherapy [15,22]. 172 These viruses are unable to replicate in healthy cells, thus providing for a treatment option that can 7 173 attack cancerous cells to the exclusion of normal cells—an advantage over existing chemotherapy 174 or radiation therapy [22,23]. This is a relatively new area of research, and largely experimental. 175 Nevertheless, many potent viruses have been designed which have performed very well in clinical 176 trials, and some have been approved onto the market. The oncolytic virus Oncorine used in 177 nasopharyngeal cancer, for example, is a type of adenovirus that has been engineered to lack the 178 E1B protein, which is responsible for deactivating the p53 protein in the host cell [23]. The tumour 179 suppressor p53 plays a crucial role in the host cell’s ability to destroy the virus, and without viral 180 E1B to deactivate the host’s p53 defense, the host’s normal cells will be able to clear the Oncorine 181 infection. However, many cancerous cells have mutations in their p53 gene which renders the 182 protein dysfunctional (a main cause of neoplastic proliferation), and so the Oncorine virus can 183 replicate in them and cause toxicity/death to the exclusion of normal cells [23]. Phase III trials of 184 the drug showed a response rate of 80% for head and neck tumours, which was double the rate of 185 40% in patients given standard chemotherapy, and other studies have consistently demonstrated a 186 good safety profile with only flu-like side effects [24]. 187 There are a number of important limitations currently facing this field. Firstly, the classes 188 of viruses from which these oncolytic agents are derived from are fairly common in nature, and 189 many humans have been exposed to and developed acquired immunity against them. Some 190 possible solutions include the use of immunosuppressants to temporarily block the immune 191 reaction, or using carrier cells to deliver the viruses directly to the tumour [23]. Moreover, for the 192 virus to be able to effectively reduce a tumour, the rate of viral replication in infected cells must 193 be greater than the growth rate of the uninfected cancerous cells. As such, this approach may not 194 be suitable for very large or fast-growing tumours. Nevertheless, the maturation of this tool will 8 195 allow for highly-specific treatments against many other forms of cancer without compromising 196 healthy tissue, and so is a promising cancer treatment of the future [15]. 197 CRISPR/Cas9 198 There is major leap forward currently underway brought by the advent of the CRISPR- 199 Cas9 tool that promises to revolutionize the entire field of gene therapy. This technology allows 200 for precise modification of DNA sequences in an efficient and simple manner, and may offer 201 several notable therapeutic uses. It is one of the fastest growing areas of gene therapy research, 202 generating a fair deal of excitement—and controversy [25]. 203 In the context of cancer research, the tool is also showing remarkable success against viral 204 infections which are implicated with the development of cancer. A recent study programmed a 205 Cas9 construct to specifically target and cleave the E6 oncogene of human papilloma virus (HPV) 206 in cervical cancer cells, and found a substantial reduction in HPV viral-load and restoration of 207 normal apoptotic genes [26]. Administration of a similar construct in HPV infected mice with 208 cervical cancer found a significant reduction in tumour size [27]. This is not without limitations 209 however, as viruses have the capacity to quickly evolve resistance against CRISPR-Cas9 210 constructs, indicating the need for more research to minimize this resistance [28]. More generally, 211 the biggest barrier to success currently is the difficulty in creating delivery methods which can 212 modify a sufficient number of cells to produce clinical benefits. This is a decidedly active area of 213 CRISPR research, and advances here are required before the tool can ever be used clinically [25]. 214 The ease and accuracy of DNA modification allowed by the tool can also be used to repair 215 deleterious mutations or replace sections of DNA with any desired sequence [25,29,30]. 216 Accordingly, the possibilities of restoring mutations accumulated in cancerous cells or fixing 217 alleles associated with increased cancer risk have been suggested as ways to control the 9 218 development of disease [25,29,30]. As a result, ethical, moral, and legal questions result from the 219 ability to modify the genetic content of human beings [29–32]. Current issues include the extent 220 to which traits and parts of the genome should be allowed for modification, the consequences of 221 socioeconomic access disparities on creating biological inequalities, and the question of whether 222 human embryos can be modified as a means to cure genetic components of disease [29–32]. The 223 capacity to “improve” humanity has also raised the specter of eugenics, which has provoked 224 considerable controversy as well as opposition from several religious, philosophical, and legal 225 perspectives [31,32]. The developers of CRISPR-Cas9 have even requested a ban on all attempts 226 at human germline modification until society can have a discussion about its consequences [30]. 227 Conclusion 228 Genetic therapy and modification is an exciting new paradigm of personalized medicine, 229 allowing for medical procedures that can target diseases such as cancer in novel ways. The 230 development of gene transfer treatments, immunotherapy, oncolytic virotherapy, and direct gene 231 editing are emerging as strong therapeutic applications (Table 1). Nonetheless, there are still many 232 questions of safety, efficacy, and commercial viability which remain to be resolved, as well as the 233 emerging controversy over the ethical, legal, and moral implications that human genetic 234 modification will have on society. These issues must be confronted and addressed through prudent 235 solutions and regulatory/legal frameworks before gene therapy and genome modification can 236 become widely available treatment modalities for human diseases [32]. 237 238 239 240 10 241 242 Table 1. Summary outlining the mechanism, advantages, and limitations of each of the classes of gene therapy modalities for cancer treatment. Class Mechanism Advantages Limitations Gene Transfer Treatments Insertion of foreign genes into target cells, mainly through viral based vectors. Other non-viral delivery methods are being developed as well. Can alter tumor cell function, restore apoptotic or tumour suppression pathways, and enable targeted disruption of specific types of cancer cells. Immunotherapy Modifies cancer cells to produce antigenic and immunostimulatory molecules, or to modify immune cells to express cytokines and T-Cell Receptors against specific tumor antigens. Produces genetically modified viruses which target and attack tumour cells preferentially over normal cells. Creates a more aggressive and targeted immune response toward tumours, Can create ‘cancer vaccines’ that are personalized for the patients tumour type and immunological environment. There are difficulties with achieving efficient transfer methods, host immune responses to many viral vectors, and concerns about safety due to increased risks of mutations. Very time-intensive and expensive; some cost estimates exceed USD 75,000. Oncolytic Virotherapy Genome Modification Tools like CRISPR/Cas9 can be used to edit or replace sections of the patient’s genome in a highly accurate and practical manner. Allows for treatment directed specifically against cancer cells with minimal side effects. Also enables targeting of certain cancers that do not respond to well to standard therapy. Can remove latent cancercausing viral infections such as HPV. Also, it is able to repair deleterious mutations, restore tumour suppression and apoptosis, and change alleles which increase cancer risk. Host can develop immune response against the viruses. Also not efficacious against fast-growing tumours due to growth rates that exceeds viral replication rate. Current delivery methods of Cas9 constructs have difficulties with efficiency, and oncogenic viruses can develop resistance against the constructs. There are also concerns over the ethics and effects of permanent genome modification. 243 244 REFERENCES 245 246 247 1. Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010;127(12):2893–917. 248 2. Varmus H. The new era in cancer research. Science. 2006;312(5777):1162–5. 249 250 3. Siegel R, Desantis C, Virgo K, Stein K, Mariotto A, Smith T, et al. Cancer Treatment and Survivorship Statistics, 2012. CA-Cancer J Clin. 2013;62(4):220–41. 251 252 4. Fioretti D, Iurescia S, Fazio VM, Rinaldi M. DNA vaccines: Developing new strategies against cancer. J Biomed Biotechnol. 2010;2010:1–16. 253 5. Cross D, Burmester JK. Gene therapy for cancer treatment: past, present and future. Clin 11 254 Med Res. 2006;4(3):218–27. 255 256 257 6. Hermonat P, Muzyczka N. Use of adeno-associated virus as a mammalian DNA cloning vector: transduction of neomycin resistance into mammalian tissue culture cells. PNAS. 1984;81(20):6466–70. 258 259 260 7. Li Y, Li B, Li C, Li L. Key points of basic theories and clinical practice in rAd-p53 (Gendicine) gene therapy for solid malignant tumors. Expert Opin Biol Ther. 2015;15(3):437–54. 261 262 8. Gordon E, Hall F. Rexin-G, a targeted genetic medicine for cancer. Expert Opin Biol Ther. 2010;10(5):819–32. 263 264 265 266 9. Chawla SP, Chua VS, Fernandez L, Quon D, Blackwelder WC, Gordon EM, et al. Advanced phase I/II studies of targeted gene delivery in vivo: intravenous Rexin-G for gemcitabine-resistant metastatic pancreatic cancer. Mol Ther. Nature Publishing Group; 2010;18(2):435–41. 267 268 10. Fischer A, Hacein-bey-abina S, Cavazzana-calvo M. Gene therapy for primary adaptive immune deficiencies. J Allergy Clin Immunol. 2011;127(6):1356–9. 269 270 271 272 11. Ohlfest JR, Demorest ZL, Motooka Y, Vengco I, Oh S, Chen E, et al. Combinatorial antiangiogenic gene therapy by nonviral gene transfer using the Sleeping Beauty transposon causes tumor regression and improves survival in mice bearing intracranial human glioblastoma. Mol Ther. 2005;12(5):778–88. 273 274 12. Patil S, Rhodes D, Burgess D. DNA-based therapeutics and DNA delivery systems: a comprehensive review. AAPS J. 2005;7(1):E61–77. 275 276 277 13. Zinn E, Pacouret S, Khaychuk V, Turunen HT, Carvalho LS, Andres-Mateos E, et al. In silico reconstruction of the viral evolutionary lineage yields a potent gene therapy vector. Cell Rep. 2015;12(6):1056–68. 278 279 14. Armstrong AC, Eaton D, Ewing JC. Cellular immunotherapy for cancer. BMJ. 2001;323(7324):1289–93. 280 281 15. Nawrocki S, Wysocki P, Mackiewicz A. Genetically modified tumour vaccines: an obstacle race to break host tolerance to cancer. Expert Opin Biol Ther. 2001;1(2):193–204. 282 283 16. Hege K, Jooss K, Pardoll D. GM-CSF gene-modifed cancer cell immunotherapies: of mice and men. Int Rev Immunol. 2006;25(5-6):321–52. 284 285 17. Eager R, Nemunaitis J. GM-CSF gene-transduced tumor vaccines. Mol Ther. 2005;12(1):18–27. 286 287 288 18. Lipson E, Sharfman W, Chen S, McMiller T, Pritchard T, Salas J. Safety and immunologic correlates of Melanoma GVAX, a GM-CSF secreting allogeneic melanoma cell vaccine administered in the adjuvant setting. J Transl Med. 2015;13:214–38. 12 289 290 19. Rosenberg S. Raising the bar: the curative potential of human cancer immunotherapy. Sci Transl Med. 2012;4(127):127ps8. 291 292 293 20. Robbins PF, Morgan RA, Feldman SA, Yang JC, Sherry RM, Dudley ME, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol. 2011;29(7):917–24. 294 21. Russell S, Peng K, Bell J. Oncolytic virotherapy. Nat Biotechnol. 2012;30(7):658–70. 295 296 297 22. Guo Z, Thorne S, Bartlett D. Oncolytic virotherapy: molecular targets in tumor-selective replication and carrier cell-mediated delivery of oncolytic viruses. Biochim Biophys Acta. 2008;1785(2):217–31. 298 299 23. Garber K. China Approves World’s First Oncolytic. J Natl Cancer Inst. 2006;98(5):298– 300. 300 301 24. Barrangou R, May AP. Unraveling the potential of CRISPR-Cas9 for gene therapy. Expert Opin Biol Ther. 2014;15(03):1–4. 302 303 304 25. Yu L, Wang X, Zhu D, Ding W, Wang L, Zhang C, et al. Disruption of human papillomavirus 16 E6 gene by clustered regularly interspaced short palindromic repeat/Cas system in human cervical cancer cells. Onco Targets Ther. 2014;8:37–44. 305 306 26. Platt R, Chen S, Zhou Y. CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell. 2014;159(2):440–55. 307 308 309 27. Wang Z, Pan Q, Gendron P, Zhu W, Guo F, Cen S, et al. CRISPR/Cas9-Derived Mutations Both Inhibit HIV-1 Replication and Accelerate Viral Escape. Cell Rep. The Authors; 2016;15:1–9. 310 311 28. Doudna J, Charpentier E. The new frontier of genome engineering with CRISPR-Cas9. Science. 2014;346(6213):1258096. 312 313 29. Lokody I. Genetic therapies: Correcting genetic defects with CRISPR Cas9. Nat Rev Genet. 2014;15(2):63. 314 315 316 30. Baltimore D, Berg P, Botchan M, Carroll D, Charo R, Church G, et al. A prudent path forward for genomic engineering and germline gene modification. Science. 2015;348(6230):36–8. 317 318 31. Lanphier E, Urnov F, Haecker S, Werner M, Smolenski J. Don’t edit the human germ line. Nature. 2015;519(7544):410–1. 319 320 32. Ishii T. Potential impact of human mitochondrial replacement on global policy regarding germline gene modification. Reprod Biomed Online. 2014;29(2):150–5. 321 13