Download Research and the Changing Landscape of Oncology: Cancer Therapy

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
Research and the Changing Landscape
of Oncology: Cancer Therapy
Yiliang Zheng
Monash University, 5th Year
Cancer Council Australia 2015 Essay Competition
Yiliang Zheng, Monash University, 5th Year
Page | 1
Essay Outline
Introduction………………………………………………………………..…………………....3
Past Highlights………………………………………………………………………………….4
Present Trends………….…………………………………………………………...………….5
Future Directions………………………………………………………………………………..7
Relevance of Research Advances and Novel Treatments to Australia Cancer Care…..……….8
Relevance of Research Advances and Novel Treatments to Medical Students – Our Future
Healthcare Professionals……………………………………………………………………...10
Conclusion………………………………...………………………………………………...…11
References………………………...…………………………………………………………..12
Yiliang Zheng, Monash University, 5th Year
Page | 2
Introduction
In 2013, Science magazine, a leading American scientific journal, proclaimed cancer
immunotherapy as the ‘Breakthrough of the Year’ and went as far as claiming it the ‘turning
point in cancer’.1 The prominent leading example of immunotherapy, ipilimumab (Yervoy), an
anti-CTLA-4 antibody, has been touted as revolutionary in the treatment of metastatic
melanoma, resulting in significantly increased overall survival.2,3 Success of ipilimumab in
melanoma opened doors to the potential use of immune checkpoint inhibitors in other
malignancies, setting the stage for many current immunotherapy research and trials.4-6
Ipilimumab’s approval of use by the US FDA in 2011 arrived a mere decade after the
approval of imatinib (Glivec), a kinase inhibitor, in 2001 which transformed the treatment of
chronic myeloid leukaemia (CML), producing complete cytogenetic response rate of 76.2%
and progression-free survival of 96.7% at 18 months.7 With the advent of imatinib, a new
wave of small-molecule therapeutics, particularly the tyrosine-kinase inhibitors (TKIs),
redefined oncologic treatment, with a shift away from broadly-acting cytotoxic agents to
more tumour-specific targeted therapies.7-9
These paradigm shifts in the oncologic treatment landscape evolved from rapid advances in
global cancer research over recent decades which improved our understanding of cancer
biology. These research breakthroughs place us at an exciting, defining moment in the
oncology timeline as we enter an era of personalized cancer medicine with immunotherapy
and targeted therapy taking centre stage in cancer research and therapeutics today.10
This essay discusses past, present and future trends in cancer therapy, highlighting the pivotal
role research plays in their development. It delves into the implications of these research and
treatment advances in the context of Australia’s cancer care and health system. It also
explores the relevance of these therapeutic and research advances to medical students.
Yiliang Zheng, Monash University, 5th Year
Page | 3
Past Highlights
The early-to-mid 20th century saw the explosion of numerous chemotherapeutic discoveries,
such as antifolates, thiopurines, vinca alkaloids, which largely relied on large-scale screening
of plants, fungi and organisms for compounds with anti-cancer activity.11-13 It was the
landmark discovery of the effectiveness of nitrogen mustard against Hodgkin’s lymphoma
post-World War II which represented the dawn of oncology.14 Although many
chemotherapeutic agents were available, most were limited by narrow therapeutic index,
toxicities and acquired resistance.11,12 Little was known about cancer biology then, though
subsequent development of chemotherapy and radiotherapy regimens was guided by the
hypothesis of tumour cells’ sensitivity to DNA damage.15
Despite subsequent discoveries of the first proto-oncogenes and tumour suppressor genes in
1970s, the first mutated gene in human cancer in 1982, and tumour signalling pathways in
1990s, genetic understandings had minor impact on clinical efforts to control cancer.15-17 Drug
development continued with compound modifications and large-scale screening, without
learning from advances in cancer biology.
Surgery and radiotherapy had always been and are likely to remain the bulk of cancer
treatment.15 Improvements in technologies in recent decades, such as minimally-invasive
surgery, intensity-modulated radiation therapy (IMRT) and image-guided radiation therapy
(IGRT) that deliver precise radiation to the target in 3-dimensions and motion respectively,
make them more effective therapies, independent to molecular advances.15,18,19
Yiliang Zheng, Monash University, 5th Year
Page | 4
Present Trends
The progress of research in recent decades that brought increasing understanding of the
molecular mechanisms of cancer pathogenesis has translated to the discovery, development
and application of molecularly-targeted agents (MTAs).20 MTAs inhibit specific molecular
pathways enabling key capabilities or hallmarks in tumour cells (such as growth and
maintenance), activate apoptotic signaling and facilitate effector functions of immune cells,
effectuating rapid and profound tumour regression.12,15,21 Following successes of imatinib in
CML and rituximab in non-Hodgkin lymphoma, emergence of these mechanism-based
therapies has changed many metastatic solid tumours from diseases with nearly invariable,
early fatality, to chronic diseases with prolonged progression-free survival (PFS) and overall
survival.22 For instance, sunitinib, a multi-targeted TKI demonstrating longer PFS than
previously used interferon-α, became the most commonly used first-line agent in metastatic
renal cell carcinoma (RCC) which, with the use of other second-line agents including everolimus
and axitinib, improved overall survival from several months to years.22-25
Parallel to the development of MTAs is the advancement in genomics and proteomics. We are
only beginning to appreciate that chromosomal and genetic defects modify cellular signaling,
creating survival advantage for tumour cells.26 Following the footsteps of the Human Genome
Project which identified more than 290 cancer-related genes through collaborative research,
the Cancer Genome Atlas (CGA) and the International Cancer Genome Consortium (ICGC) are
monumental international efforts to provide a complete catalogue of all cancer genes in major
cancer types.27-29 Through genetic sequencing and molecular characterization of hundreds of
tumours, they aim to provide a comprehensive description of all genomic, transcriptomic and
epigenomic changes.29,30
Knowledge of oncogenic mutations and molecular changes holds promise to reveal potential
therapeutic targets and develop MTAs to treat specific patients’ tumours. Current research
identifies oncoproteins in various signaling pathways involved in tumour growth and
maintenance present in patient subpopulations, such as EGFR, HER2, BRAF, c-KIT – these act as
predictive biomarkers that predict response to specific MTAs.12,31-33 Molecular profiling of
patients’ tumours performed prior to treatment can determine their mutational status which can
be matched to particular therapeutic agents.31 For example, EGFR inhibitors cetuximab and
panitumumab are used for patients with KRAS mutation-negative colorectal carcinomas,
extending their PFS when combined with chemotherapy.15,22 Gefitinib, another EGFR inhibitor,
was used in lung adenocarcinoma expressing EGFR-activating mutation, demonstrating
significant PFS in the IPASS study in 2004.15,22,32,34 Trastuzumab, a HER2-inhibitor, is used as
part of the adjuvant therapy for HER2-overexpressing breast cancer.15,22 Presence of BRAF
V600E mutations in metastatic melanoma predicts favourable use of vemurafenib, an antiBRAF V600E antibody.15,22 With our shift towards precision medicine, molecular heterogeneity
of cancers means targeted therapies based on individual molecular aberrations will be
increasingly developed and applied in cancer therapy.35
The emergence of novel immune-based strategies came with an expansion in understanding of
immune controls and responses to tumours, particularly immune escape and checkpoint
blockade of inhibitory receptors and ligands, such as CTLA-4, PD1/PDL-1.36,37 Dr Jim Allison’s
Yiliang Zheng, Monash University, 5th Year
Page | 5
research in immune responses and T cells led to the discovery of ipilimumab and its subsequent
use in metastatic melanoma.37,38 By stimulating host immune responses, ipilimumab and other
immunotherapeutics, including cell vaccines, cytokines and anti-PD1 antibodies, have
demonstrated extraordinary clinical efficacy with durable tumour regression, especially in
diseases known to respond to immune-based therapies, like melanoma and RCC.37,39,40
Immunotherapy is showing promise in treating traditionally non-responding diseases and
current trials seek to further evaluate response rates.4-6,36,37,41 For example, pembrolizumab
and nivolumab, two anti-PD-1 antibodies approved for melanoma, are currently evaluated in
solid tumours such as non-small-cell lung cancers, breast and bladder cancers.41-43
Yiliang Zheng, Monash University, 5th Year
Page | 6
Future Directions
Targeted therapy and immunotherapy, with the emerging molecular and genetic
characterization of cancers, pose an alluring revolution in cancer treatment. However, much
work remains to be done before these novel therapeutics hold dominance in cancer treatment
of tomorrow.
Comprehensive catalogues of cancer genes in the CGA and ICGC need to be completed while
efficient, extensive methods of molecular characterization and genomic sequencing of tumours
have to be developed.30,31,35 We need to develop sensitive and specific predictive
biomarkers which match genomic abnormalities and predict response to MTAs; they will help
stratify patients that may benefit from specific MTAs.11,44 Clinical efficacy and safety of MTAs
need further evaluation.20,45 Drug resistance, especially with MTAs, is a pertinent problem and
novel strategies are required to overcome such resistance.15 We will need to deepen our
understanding of the immune system to uncover components that can be harnessed with
immunotherapy.46 We can optimize targeted therapies and immunotherapies, expand their
applications and evaluate their roles in adjuvant and neo-adjuvant settings.12,47-53 This allows
us to ultimately establish combined-multi-modal therapies for the individual patient to produce
rapid responses with durable remissions.12,47-53
These evaluations will require intensive research and trials to advance the roles these novel
therapies play in current and future mainstream cancer therapy. With the redefinition and
classification of cancers based on genomics, personalized, precise cancer therapy using
multimodal agents based on tumour-specific molecular profiles will likely be the standard
oncologic practice of tomorrow.
With emerging innovations in specialized fields, multiple treatment modalities and approaches
which may potentially be employed in future cancer therapy are being explored.
Investigations are underway in epigenomic engineering where cancer expression can be
modulated with precisely reversed epigenetic mutations through genomic editing and
reprogramming.54,55 Therapeutic radioisotopes with antibodies targeting tumour components
are developed in radioimmunotherapy while nanoparticles are explored as delivery vehicles
of therapeutics into tumour cells.56-61 Novel therapeutic targets such as miRNA, mitochondria
and cancer stem cells are investigated. Modulating miRNA expression may inhibit tumour
proliferation as miRNA are postulated to be involved in tumourigenic processes and regulating
resistance mechanisms.62-64 Mitochondrial-targeting agents can possibly suppress tumour
metastasis by modulating mitochondrial function and oxidation.65-68 Cancer stem cells are
increasingly recognized as potential treatment targets with strategies evaluated to inhibit their
pathways.69
Yiliang Zheng, Monash University, 5th Year
Page | 7
Relevance of Research Advances and Novel Treatments to Australia
Cancer Care
Australia is no stranger to the devastating impact of cancer which was estimated to be its
leading cause of burden of disease in 2012 with rising incidence rates.70,71 With the global
revolution in novel therapeutics and surge in genetic understanding of cancer, Australia has
both contributed to and benefited from these recent advances in research and therapy.
Australia’s scientific and medical communities actively contribute to the oncologic scene by
continually expanding knowledge of cancer biology and establishing trials for new drugs.
Pancreatic cancer is Australia’s major contribution to the ICGC through the Australian
Pancreatic Cancer Genome Initiative.72 By performing whole-genome sequencing and copy
number variation analysis of 100 pancreatic ductal adenocarcinomas, research teams
uncovered genetic drivers in tumourigenesis and highlighted 4 subtypes with potential clinical
utility.72-74 This allows investigators to match mutations to specific MTAs and test their efficacy
on patient subpopulations.73 The Australia and New Zealand Breast Cancer Trials Group
(ANZBCTG) continuously conducts trials that provided the most reliable evidence supporting
use of many treatment strategies in breast cancer, including tamoxifen and combination
therapy.75 In collaboration with other international research groups, such as the Breast Cancer
International Research Group, ANZBCTG seeks to further evaluate the use of specific targeted
therapies.75 The Melanoma Institute Australia similarly conducts various clinical trials to
evaluate efficacy of novel therapies in melanoma.76
While excitement mounts in Australia with the new wave of oncology medicines as prospective
therapies, their arrival poses challenges to regulatory and reimbursement processes that may
limit access to these medicines.
In Australia, drugs have to be approved for use by the Therapeutic Goods Administration
(TGA) and for subsidy on the Pharmaceutical Benefits Scheme (PBS) as recommended by the
Pharmaceutical Benefits Advisory Committee.77,78 The PBS is currently the only means of
providing Australians with broad and equitable access to cancer medicines.79 Unfortunately,
there is often a prolonged PBS approval process for new cancer medicines.79 Many Australian
patients cannot wait for the subsidy approval nor afford the full unsubsidized cost to access
new drugs.80 An illustrative example is in melanoma, with highest world-wide incidence in
Australia where the arrival of ipilimumab was highly anticipated.81 Ipilimumab was registered
by the TGA in 2011 for use in metastatic melanoma, but costs AUD$190,341.20 for 4
injections.82,83 Subsequent inclusion of ipilimumab on the PBS in 2013 allowed Australians to
only pay AUD$150.80.83,84
Subsidizing cancer medicines is costly. Expenditure on cancer medicines by the PBS has
substantially grown more than 60% in the five years to 2011, costing $587.5 million in
2012.85 Many stakeholders question the appropriateness of funding these expensive
medicines which may translate to marginal benefits for selected patients, diverting funds from
many other competing priorities.86
Yiliang Zheng, Monash University, 5th Year
Page | 8
Physical access to novel therapeutics is an additional issue in Australia. A third of Australians
with cancer live in rural-regional areas with poor access to cancer treatment and, consequently,
face significantly poorer survival outcomes, having up to 300% higher five-year mortality risk
for certain cancers.87,88 With the revolution of novel therapies, the gap of cancer burden in
Australia widens with patients in rural areas bearing a larger disproportionate part of the
burden as new therapies are primarily restricted to urban-based tertiary centres.
These challenging issues of timely, affordable and geographical access remain difficult to
resolve, expecting to worsen with the burgeoning surge in novel therapeutics. To date, the
Australian government remains supportive of continued listing of cancer drugs on the PBS,
recognizing their necessity in cancer treatment, while implementing measures to curb
expenditure growth.89 Establishment of the Rural Cancer Centres Initiative and patient-assisted
travel schemes may reduce geographical discrepancy in cancer care outcomes by improving
rural-regional access to cutting-edge therapies.88
Yiliang Zheng, Monash University, 5th Year
Page | 9
Relevance of Research Advances and Novel Treatments to Medical
Students – Our Future Healthcare Professionals
As cancer survival rates improve, more people are living with cancer today. On average, GPs
are said to encounter, annually, 4 new patients diagnosed with a potentially fatal cancer and
have, at any one time, 16 patients with cancer.90 Regardless of the discipline that medical
students eventually practice in, they will be expected to manage oncology patients to varying
degrees. It is hence important for students to have a firm and comprehensive understanding of
cancer management, as outlined in the Ideal Oncology Curriculum published by the Cancer
Council Australia, including established treatments of surgery, radiotherapy and
chemotherapy.91
Most common and several rare malignancies have at least a systemic therapy, such as an MTA
or immunotherapy, and doctors will encounter patients receiving these novel therapies.92 As
use of newer therapeutic agents become more widespread across disciplines, all healthcare
professionals will need to be familiar with them as they become the mainstay of treatment.92
Medical students today should ideally be aware of them and learn their side-effects as part
of medical school oncology education.
Medical students should also be equipped with knowledge in genetics and genome-wide
studies and skills in evidence-based medicine. This will allow them to effectively critically
appraise and contribute to cancer research.
Yiliang Zheng, Monash University, 5th Year
Page | 10
Conclusion
In over half a century, we have made great strides in cancer therapy with refined treatments
and improved five-year survival rates. As we witness the ever-changing landscape of
oncologic therapy, from surgery, radiotherapy and chemotherapy, to the advent of MTAs and
immunotherapy, it is easy to overlook the work of research that drives and inspires these
paradigm shifts to give us the armamentarium in cancer treatment of today. This progress is
due to the pooling of expertise of individuals with specialized skills working collaboratively in
multidisciplinary teams and convergence of different fields with independent research threads.
Each clinician and researcher contributes through small incremental advances to the lattice of
knowledge constructed over decades, allowing remarkable breakthroughs to produce novel
therapies. We need to maintain concerted international efforts in all aspects of modern
oncology – from research, drug development, and clinical application of novel agents – to
facilitate the effective translation of cancer biology to therapeutic benefits for cancer patients.
As we stand at the dawn of a new era of cancer research and therapy, we anticipate radical
changes to the future landscape. Each wave of discovery will bring new insights, challenges,
renewed hope and emerging, promising treatments. Our ceaseless hunt for ever-better
treatment will continuously push frontiers of research with ever-expanding understanding of
cancer biology and novel therapies.
Yiliang Zheng, Monash University, 5th Year
Page | 11
References
1. Cancer Research Institute. Cancer immunotherapy named 2013 “Breakthrough of the Year”
[Internet]. New York: Cancer Research Institute; 2013 [updated 2013 Dec 23, cited 2015
Mar 15]. Available from: http://www.cancerresearch.org/news-publications/ourblog/december-2013/cancer-immunotherapy-named-2013-breakthrough-of-the-year.
2. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved
survival with ipilimumab in patients with metastatic melanoma. N Engl J
Med. 2010;363:211–223.
3. Maio M, Grob JJ, Aamdal S, Bondarenko I, Robert C, Thomas C, et al. Five-year survival
rates for treatment-naive patients with advanced melanoma who received ipilimumab plus
dacarbazine in a phase III trial. Journal of Clinical Oncology. 2015 Feb 23. Pii:
JCO.2014.56.6018.
4. Pilotto S, Kinspergher S, Peretti U, Calio A, Carbognin L, Ferrara R, et al. Immune
checkpoint inhibitors for non-small-cell lung cancer: does that represent a ‘new frontier’?
Anticancer Agents Med Chem. 2015;15(3):307-13.
5. Domingues D, Turner A, Silva MD, Marques DS, Mellidez JC, Wannesson L, et al.
Immunotherapy and lung cancer: current developments and novel targeted therapies.
Immunotherapy. 2014;6(11):1221-35.
6. Luke JJ, Ott PA. PD-1 pathway inhibitors: the next generation of immunotherapy for
advanced melanoma. Oncotarget. 2015 Feb 28;6(6):3479-92.
7. O’Brien SG, Guilhot F, Larson RA, Gathmann I, Baccarani M, Cervantes F, et al. Imatinib
compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase
chronic myeloid leukaemia. N Engl J Med. 2003 March;348(11):994-1004.
8. Van’t Veer LJ, Bernards R. Enabling personalized cancer medicine through analysis of
gene-expression patterns. Nature. 2008 Apr 3;452(7187):564-70.
9. Schilsky RL. Personalised medicine in oncology: the future is now. Nature Reviews Drug
Discovery. 2010 May;9:363-366
10. Joseph SO, Wu J, Muggia FM. Targeted therapy and its status and promise in selected
solid tumours part II: impact on selected tumour subtsets and areas of evolving integration.
Oncology. 2012 Nov;26(11):1021-30.
11. Zaenker KS and Entschladen F. Paving roads for new drugs in oncology. Recent Pat
Anticancer Drug Discov. 2009 Jun;4(2):137-145.
12. Vanneman M, Dranoff G. Combining immunotherapy and targeted therapies in cancer
treatment. Nature Reviews Cancer. 2012 Apr;12:237-251.
13. DeVita VT Jr, Chu E. A history of cancer chemotherapy. Cancer Res. 2008 Nov 1;68:8643
14. Goodman L, Wintrobe M, Dameshek W, Goodman M, Gilman A, McLennan M. Nitrogen
mustard therapy. Use of methyl-bis(beta-chloroethyl)amine hydrochloride and tris(betachloroethyl)amine hydrochloride for Hodgkin’s disease, lymphosarcoma, leukemia and
certain allied and miscellaneous disorders. JAMA. 1946;251(17):2255-61.
15. Varmus H. The New Era in Cancer Research. Science. 2006 May 26;312(5777):11621165.
16. Reddy EP, Reynolds RK, Santos E, Barbacid M. A point mutation is responsible for the
acquisition of transforming properties by the T24 human bladder carcinoma oncogene.
Nature. 1982 Nov 11;300(5888):149-52.
Yiliang Zheng, Monash University, 5th Year
Page | 12
17. Futreal PA, Coin L, Marshall M, Down T, Hubbard T, Wooster R, et al. A census of human
cancer genes. Nat Rev Cancer. 2004 Mar;4(3):177-83.
18. UC San Diego School of Medicine. History of radiation therapy [Internet]. La Jolla CA: UC
San Diego; 2015 [updated 2015; cited 2015 Mar 2]. Available from:
http://healthsciences.ucsd.edu/som/radiation-medicine/about/Pages/history-radiationtherapy.aspx.
19. Cleveland Clinic. Image guided radiation therapy [Internet]. Cleveland Ohio: Cleveland
Clinic; 2015 [updated 2015; cited 2015 Mar 2]. Available from:
http://my.clevelandclinic.org/services/cancer/treatments-procedures/radiationtherapy/image-guided-radiation-therapy.
20. Venkatakrishnan K, Friberg LE, Ouellet D, Mettetal J, Stein A, Troconiz I, et al. Optimising
oncology therapeutics through quantitative translational and clinical pharmacology:
challenges and opportunities. Clin Pharmacol Ther. 2015 Jan;97(1):37-54.
21. Ribas A and Wolchok JD. Combining cancer immunotherapy and targeted therapy.
Current Opinion in Immunology. 2013 April;25(2):291-296.
22. Wu J, Joseph SO, Muggia FM. Targeted therapy and its status and promise in selected
solid tumours part I: areas of major impact. Oncology. 2012 Oct;26(10):935-43.
23. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon alfa in metastatic renalcell carcinoma. N Engl J Med. 2007;356:115-24.
24. Morias C. Sunitinib resistance in renal cell carcinoma. Journal of Kidney Cancer and VHL.
2014;1(1):1-11
25. Singh P, Agarwal N, Pal SK. Sequencing systemic therapies for metastatic kidney cancer.
Curr Treat Options Oncol. 2015 Jan;16(1):316.
26. Hanahan D and Weinberg RA. Hallmarks of Cancer: The Next Generation. Cell. 2011
March 4;144(5):646-674.
27. American Association for Cancer Research. AACR cancer progress report: progress in
cancer research [Internet]. Philadelphia PA: American Association for Cancer Research;
2011 [updated 2012; cited 2015 Mar 20]. Available from:
http://cancerprogressreport.org/2011/Pages/ProgressInCancerResearch.aspx?Page=1.
28. National Cancer Institute. Analysis using TCGA data identifies therapeutic possibility
[Internet]. Bethesda MD: National Cancer Institute; 2013 [updated 2013 Jun 18; cited
2015 Mar 22]. Available from:
http://cancergenome.nih.gov/researchhighlights/researchbriefs/Analysis_Using_TCGA_D
ata_Identifies_New_Therapeutic_Possibility.
29. International Cancer Genome Consortium. ICGC cancer genome projects [Internet].
International Cancer Genome Consortium; 2015 [updated 2015 Jan 21; cited 2015 Mar
22]. Available from: https://icgc.org/.
30. Lawrence MS, Stojanov P, Mermel CH, Robinson JT, Garraway LA, Golub TR, et al.
Discovery and saturation analysis of cancer genes across 21 tumour types. Nature. 2014
Jan 23;505(7484):495-501.
31. Doroshow JH and Kummar S. Translational research in oncology – 10 years of progress
and future prospects. Nature Reviews Clinical Oncology. 2014 Nov;11(11):649-662.
32. John T, Liu G, Tsao MS. Overview of molecular testing in non-small-cell lung cancer:
mutational analysis, gene copy number, protein expression and other biomarkers of EGFR
for the prediction of response to tyrosine kinase inhibitors. Oncogene. 2009;28:S14-S23.
Yiliang Zheng, Monash University, 5th Year
Page | 13
33. Dienstmann R, Rodon J and Tabernero J. Drug development in the era of personalised
oncology: from population-based trials to enrichment and prescreening strategies. Am Soc
Clin Oncol Educ Book. 2012:168-72.
34. Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijio N, et al. Gefitinib or
carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361:947-57.
35. Akkad J, Bochum S, Martens UM. Personalised treatment for colorectal cancer: novel
developments and putative therapeutic strategies. Langenbecks Arch Surg. 2015
Feb;400(2):129-43.
36. Rolfo C, Sortino G, Smits E, Passiglia F, Bronte G, Castiglia M, et al. Immunotherapy: is a
minor god yet in the pantheon of treatments for lung cancer? Expert Rev Anticancer Ther.
2014 Oct;14(10):1173-87.
37. Eggermont Am, Kroemer G, Zitvogel L. Immunotherapy and the concept of a clinical cure.
Eur J Cancer. 2013 Sep;49(14):2965-7.
38. The University of Texas MD Anderson Cancer Center. Cancer pioneer James Allison leads
MD Anderson immunotherapy efforts [Internet]. Houston TX: The University of Texas MD
Anderson Cancer Center; 2012 [updated 2012 Feb 11; cited 2015 Mar 23]. Available
from: http://www.mdanderson.org/newsroom/news-releases/2012/james-allison.html.
39. Stadler S, Weina K, Gebhardt C, Utikal J. New therapeutic options for advanced nonresectable malignant melanoma. Adv Med Sci. 2014 Dec 25;60(1):83-88.
40. Gunturi A, McDermott DF. Potential of new therapies like anti-PD1 in kidney cancer. Curr
Treat Options Oncol. 2014 Mar;15(1):137-46.
41. Madureira P, de Mello RA, de Vasconcelos A, Zhang Y. Immunotherapy for lung cancer:
for whom the bell tolls? Tumour Biol. 2015 Mar 4.
42. Shin DS, Ribas A. The evolution of checkpoint blockade as a cancer therapy: what’s here,
what’s next? Curr Opin Immunol. 2015 Jan 23;33C:23-35.
43. Mahoney KM, Atkins MB. Prognostic and predictive markers for the new immunotherapies.
Oncology (Williston Park). 2014 Nov;28 Suppl 3:39-48.
44. Fuzery AK, Levin J, Chan MM, Chan DW. Translation of proteomic biomarkers into FDA
approved cancer diagnostics: issues and challenges. Clinical Proteomics. 2013;10:13.
45. Liauw WS. Molecular mechanisms and clinical use of targeted anticancer drugs. Aust Prescr
2013;36:126-31.
46. Brayer J, Fishman M. Regression of metastatic clear cell kidney cancer with interleukin-2
treatment following nivolumab (anti-PD1) treatment. J Immunother. 2014 Apr;37(3):18791.
47. Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy.
JCO. 2015 Jan 20.
48. Antonia SJ, Larkin J, Ascierto. Immuno-oncology combinations: a review of clinical
experience and future prospects. Clin Cancer Res. 2014 Dec 15;20:6258
49. Aris M, Marrio MM. Combining immunotherapy with oncogene-targeted therapy: a new
road for melanoma treatment. Front Immunol. 2015 Feb 9;6:46.
50. Rousseau B, Champiat S, Loirat D, Arrondeau J, Lemoine N, Soria JC. Immunotherapies and
targeted therapies in medical oncology. Bull Cancer. 2014 Jan 1;101(1):31-9.
51. Zheng Y, Dou Y, Duan L, Cong C, Gao A, Lai Q, et al. Using chemo-drugs or irradiation to
break immune tolerance and facilitate immunotherapy in solid cancer. Cell Immunol. 2015
Mar;294(1):54-59.
Yiliang Zheng, Monash University, 5th Year
Page | 14
52. Wargo JA, Cooper ZA, Flaherty KT. Universes collide: combining immunotherapy with
targeted therapy for cancer. Cancer Discovery. 2014 Dec;4:1377.
53. Hu-Liewskovan S, Robert L, Moreno BH, Ribas A. Combining targeted therapy with
immunotherapy in BRAF-mutant melanoma: promise and challenges. JCO. 2014 Jul
20;32(21):2248-2254.
54. Heninger E, Kruger TE, Lang JM. Augmenting autitumour immune response with epigenetic
modifying agents. Front Immunol. 2015 Feb 4;6:29.
55. Falahi F, Sgro A, Blancafort P. Epigenome engineering in cancer: fairytale or a realistic
path to the clinic? Front Oncol. 2015 Feb 6;5:22.
56. Sollini M, Boni R, Traino CA, Lazzeri E, Pasqualetti F, Modeo L, et al. New approaches for
imaging and therapy of solid cancer. Q J Nucl Med Mol Imaging. 2015 Feb 19.
57. Le VM, Wang JJ, Yuan M, Nguyen TL, Yin GF, Zheng YH, et al. An investigation of
antitumour efficacy of novel sustained and targeted 5-fluorouracil nanoparticles. Eur J
Med Chem. 2015 Mar 6;92:882-9.
58. Lin CM, Kao WC, Yeh CA, Chen HJ, Lin SZ, Hsieh HH, et al. Hyaluronic acid-fabricated
nanogold delivery of the inhibitor of apoptosis protein-2 siRNAs inhibits benzo[a]pyreneinduced oncogenic properties of lung cancer A549 cells. Nanotechnology. 2015 Mar
13;26(10):105101.
59. Wang H, Lee DK, Chen KY, Chen JY, Zhang K, Silva A, et al. Mechanism-independent
optimisation of combinatorial nanodiamond and unmodified drug delivery using a
phenotypically-driven platform technology. ACS Nano. 2015 mar 24;9(3):3332-44.
60. Estanqueiro M, Amaral MH, Conceicao J, Sousa Lobo JM. Nanotechnological carriers for
cancer chemotherapy: the state of the art. Colloids Surg B Biointerfaces. 2015 Feb
1;126C:631-648.
61. Krishnamurthy S, Ke X, Yang YY. Delivery of therapeutics using nanocarriers for targeting
cancer cells and cancer stem cells. Nanomedicine. 2015 Jan;10(1):143-60.
62. Eroles P, Tormo E, Pineda B, Espin E, Lluch A. MicroRNAs in breast cancer: one more turn in
regulation. Curr Drug Targets. 2015 Feb 12.
63. Zhang Q, Ren W, Huang B, Yi L, Zhu H. MicroRNA-183/182/96 cooperatively regulates
the proliferation of colon cancer cells. Mol Med Rep. 2015 Feb 18.
64. Costa PM, Cardoso AL, Mano M, de Lima MC. MicroRNAs in glioblastoma: role in
pathogenesis and opportunities for targeted therapies. CNS Neurol Disord Drug Targets.
2015;14(2):222-38.
65. Deus CM, Santos GL, Loureiro R, Vega-Naredo I, Faneca H, Oliveira PJ. Shutting down the
furnace: preferential killing of cancer cells with mitochondrial-targeting molecules. Curr
Med Chem. 2015 Feb 9.
66. Xu Q, Biener-Ramanuian E, Yang W, Ramanujan VK. Targeting metabolic plasticity in
breast cancer cells via mitochondrial complex I modulation. Breast Cancer Res Treat. 2015
Feb;150(1):43-56.
67. Lu J, Tan M, Cai Q. The Warburg effect in tumor progression: mitochondrial oxidative
metabolism as an anti-metastasis mechanism. Cancer Lett. 2015 Jan 28;356(2 Pt A):15664.
68. Vatrinet R, Iommarini L, Kurelac I, Deluise M, Gasparre G, Porcelli AM. Targeting
respiratory complex I to prevent the Warburg effect. Int J Biochem Cell Biol. 2015 Feb 7.
Yiliang Zheng, Monash University, 5th Year
Page | 15
69. Chiotaki R, Polioudaki H, Theodoropoulos PA. Cancer stem cells in solid and liquid tissues
of breast cancer patients: characterisation and therapeutic perspectives. Curr Cancer Drug
Targets. 2015 Feb 10.
70. Australian Institute of Health and Welfare (AIHW). Australian Cancer Incidence and
Mortality (ACIM) books: All cancers combined [Internet]. Canberra: AIHW; 2015 [updated
2015 Jan 8; cited 2015 Mar 26]. Available from: http://www.aihw.gov.au/acim-books.
71. Australian Institute of Health and Welfare & Australasian Association of Cancer Registries.
Cancer in Australia: an overview [Internet]. Canberra: AIHW; 2012 [updated 2012; cited
2015 Mar 26]. Available from:
http://www.aihw.gov.au/WorkArea/DownloadAsset.aspx?id=60129542353.
72. Garvan Institute. Pancreatic cancer research [Internet]. Australia: Garvan Institute; 2013
[updated 2013; cited 2015 Mar 26]. Available from:
http://www.garvan.org.au/research/cancer/pancreatic-cancer-research.
73. Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, et al. Whole genomes
redefine the mutational landscape of pancreatic cancer. Nature. 2015 Feb
26;518(7540):495-501.
74. International Cancer Genome Consortium. Pancreatic cancer – ductal adenocarcinoma
[Internet]. International Cancer Genome Consortium; 2015 [updated 2015 Jan 21; cited
2015 Mar 22]. Available from: https://icgc.org/icgc/cgp/68/304/798.
75. Cancer Forum. The Australian New Zealand Breast Cancer Trials Group: some contributions
to breast cancer trials [Internet]. Australia: Cancer Forum; 2009 [updated 2009 May 6;
cited 2015 Mar 26]. Available from:
http://cancerforum.org.au/Issues/2006/May/Forum/The_Australia_New_Zealand_Breast
_Cancer_Trials_Group.htm.
76. Melanoma Institute Australia. Research publications [Internet]. Australia: Melanoma Institute
Australia; 2015 [updated 2015; cited 2015 Mar 26]. Available from:
http://www.melanoma.org.au/research/research-resources/research-publications/.
77. Therapeutic Goods Administration. Australia Public Assessment Reports for prescription
medicines (AusPARs) [Internet]. Australia: Therapeutic Goods Administration; 2015
[updated 2015 Mar 20; cited 2015 Mar 31]. Available from:
https://www.tga.gov.au/australian-public-assessment-reports-prescription-medicinesauspars.
78. Pharmaceutical Benefits Scheme. PBS frequently asked questions [Internet]. Australia:
Pharmaceutical Benefits Scheme; 2015 [updated 2015 Jan 1; cited 2015 Mar 31].
Available from:
http://www.pbs.gov.au/info/general/faq#WhatisthePharmaceuticalBenefitsAdvisoryCom
mittee.
79. Cancer Drugs Alliance. Submission to the Select Committee on Health – Response [Internet].
Australia: Cancer Drugs Alliance; 2014 Sep [cited 2015 Mar 31]. 5 p.
80. Cancer Voices Australia. Submission to the Senate Inquiry into the availability of new,
innovative and specialist cancer drugs in Australia [Internet]. Australia: Cancer Voices
Australia; 2015 Feb [cited 2015 Mar 31]. 7 p. Available from:
http://www.cancervoicesaustralia.org/wp-content/uploads/2015/03/Senate-InquiryNew-Cancer-Drugs-CVA-Submision-2015.pdf.
Yiliang Zheng, Monash University, 5th Year
Page | 16
81. Alexander M, Mellor JD, McArthur G, Kee D. Ipilimumab in pretreated patients with
unresectable or metastatic cutaneous, uveal and mucosal melanoma. Med J Aust
2014;201(1):49-53.
82. Pharmaceutical Benefits Scheme. Ipilimumab, concentrate solution for I.V. infusion, 50 mg in
10mL, 200 mg in 40 mL, Yervoy – November 2012 [Internet]. Australia: Pharmaceutical
Benefits Scheme; 2012 [updated 2012 Nov; cited 2015 Apr 11]. Available from:
http://www.pbs.gov.au/info/industry/listing/elements/pbac-meetings/psd/201211/ipilimumab.
83. Pharmaceutical Benefits Scheme. Ipilimumab [Internet]. Australia: Pharmaceutical Benefits
Scheme; 2015 [updated 2015; cited 2015 Apr 11]. Available from:
http://www.pbs.gov.au/medicine/item/2638W-2641B-2643D-2663E.
84. Sunbedban.com. Ipilimumab has now been approved for PBS reimbursement in Australia
[Internet]. Australia: sunbedban.com; 2013 [updated 2013 Jul 1; cited Apr 11]. Available
from: http://sunbedban.com/ipilimumab-has-now-been-approved-for-pbs-reimbursementin-australia/.
85. Department of Health and Ageing and Medicines Australia (joint report). Trends in and
drivers of Pharmaceutical Benefits Scheme expenditure [Internet]. Australia: Department of
Health and Ageing and Medicines Australia; 2013 May [cited 2015 Mar 31]. 69 p.
Available from: http://www.pbs.gov.au/publication/reports/trends-in-and-drivers-of-pbsexpenditure.pdf.
86. Medicines Australia Oncology Industry Taskforce. Access to cancer medicines in Australia
[Internet]. Australia: Deloitte Access Economics; 2013 July [cited 2015 Mar 31]. 125 p.
Available from: https://medicinesaustralia.com.au/wpcontent/uploads/sites/52/2013/07/Access-to-oncology-medicines-1707-FINALV3.pdf.
87. National Rural Health Alliance. Cancer in rural Australia [Internet]. Canberra ACT:
National Rural Health Alliance; 2012 [updated 2012 January; cited 2015 March 20].
Available from: http://ruralhealth.org.au/sites/default/files/publications/fact-sheet-08cancer-rural-australia.pdf.
88. Cancer Council Australia. Closing the loop: bringing patients to the new regional centres
[Internet]. Australia: Cancer Council Australia; 2012 [updated 2012 Aug 7; cited 2015
Apr 7]. Available from: http://www.cancer.org.au/policy-and-advocacy/electionpriorities-2010/closing-the-loop-bringing-patients-to-the-new-regional-centres.html.
89. Parliament of Australia. Growth in expenditure on high cost drugs in Australia [Internet].
Australia: Parliament of Australia; 2015 [updated 2015 Jan 7; cited 2015 Apr 7].
Available from:
http://www.aph.gov.au/About_Parliament/Parliamentary_Departments/Parliamentary_Li
brary/pubs/rp/rp1415/ExpendCostDrugs.
90. Cancer Council Victoria. Cancer in general practice – a practical guide for primary health
care nurses [Internet]. [updated 2012 December 1; cited 2015 March 20]. Available from
http://www.cancervic.org.au/downloads/cpc/gps/cancer-care/Lesley-Pugh.pdf.
91. Sabesan S, Olver I, editors. Ideal oncology curriculum for medical schools. Sydney: Cancer
Council Australia. [Version URL:
http://wiki.cancer.org.au/oncologyformedicalstudents_mw/index.php?title=Clinical_Oncol
ogy_for_Medical_Students&oldid=1656, cited 2015 Mar 30]. Available from:
http://wiki.cancer.org.au/oncologyformedicalstudents/Clinical_Oncology_for_Medical_St
udents.
Yiliang Zheng, Monash University, 5th Year
Page | 17
92. Liauw WS. Molecular mechanisms and clinical use of targeted anticancer drugs. Aust Prescr
2013;36:126-31.
Yiliang Zheng, Monash University, 5th Year
Page | 18