touchONCOLOGY touchONCOLOGY
Gastrointestinal Cancers, Pancreatic Cancer
Read Time: 12 mins

Novel Strategies on the Horizon for Metastatic Pancreatic Cancer Management

Copy Link
Published Online: May 31st 2019 Oncology & Hematology Review. 2019;15(1):27–32 DOI: https://doi.org/10.17925/OHR.2019.15.1.27
Authors: Mehmet Akce, Bassel F El-Rayes
Quick Links:
Abstract
Article
Article Information
Abstract:
Overview

Pancreatic cancer has a dismal 5-year overall survival despite modest improvements in systemic chemotherapeutic regimens in recent years. Several new and promising agents are being tested in pancreatic cancer; these agents can be divided into three categories: immune therapy, molecular targeted agents, and stromal targeted agents. The unprecedented success of immune checkpoint inhibitors in several tumor types has not been observed in pancreatic cancer due to the unique, profoundly immunosuppressive tumor microenvironment. Despite failure of single-agent immune checkpoint inhibitors as a therapeutic option, a concerted effort is underway to further characterize the reasons for lack of response and to develop combination strategies to overcome the resistance. Molecular characterization of pancreatic cancer has led to the discovery of new molecularly-targeted therapeutic options in certain subgroups of patients, such as microsatellite instability-high tumors and BRCA-mutated tumors. Therapeutic approaches targeting DNA damage deficiency, cancer stemness, and mitochondrial energy metabolism are being tested in clinical trials. Pancreatic stroma and tumor microenvironment impact drug delivery and promote tumor growth and metastasis. Agents targeting the tumor microenvironment are being tested in combination with cytotoxic chemotherapy. This review will detail these novel agents and the encouraging results from early-phase clinical trials.

Keywords

Pancreatic cancer, immunotherapy, tumor microenvironment, DNA damage response, stromal remodeling, cancer stem cell

Article:

Pancreatic cancer is estimated to become the second most common cause of cancer-related death by 2030, and most common by 2050 in the USA.1,2 Pancreatic cancer results in approximately 331,000 deaths annually worldwide, making it the seventh most common cause of cancer-related mortality.3 More than 90% of all pancreatic cancers are pancreatic ductal adenocarcinoma (PDAC).4 For all stages combined, PDAC has a very poor 5-year overall survival (OS) rate of 8%.5 Despite significant improvements in survival rates in many cancer types, PDAC death rate has increased in recent years.5 Recent advances in chemotherapy regimens have had a modest impact on median OS, which ranges between 8.5–11.1 months.6,7 Development of novel systemic therapies to overcome the resistance of PDAC is an urgent need. This article aims to provide an in-depth review of the literature around novel treatment strategies and ongoing research in pancreatic cancer management.

Overview of current systemic treatment approach to pancreatic ductal adenocarcinoma

Single-agent gemcitabine had been the established cytotoxic chemotherapy option based on clinical benefit defined as improvement in disease-related symptoms in patients with metastatic pancreatic cancer.8 The median OS with single-agent gemcitabine is 5.6 months. Several randomized trials combining different agents with gemcitabine have failed to provide any survival benefit.9–12 However, there have been some successful gemcitabine combinations; in a double-blind international phase III study, a statistically significant OS benefit was shown by gemcitabine plus erlotinib combination compared to gemcitabine plus placebo, 6.2 versus 5.9 months.13 Significant improvements in median OS have also been observed in two randomized trials – ACCORD 11 and MPACT (Metastatic Pancreatic Adenocarcinoma Clinical Trial) – both of which established the modern treatment regimens in the front-line setting.6,7 The ACCORD 11 trial compared FOLFIRINOX regimen (leucovorin, fluorouracil [5FU], irinotecan, and oxaliplatin) to single-agent gemcitabine. Median OS was 11.1 months versus 6.8 months, respectively. The multinational MPACT trial compared gemcitabine plus nab-paclitaxel combination to single-agent gemcitabine in newly diagnosed metastatic pancreatic cancer. Median OS was 8.5 months versus 6.7 months, favoring the combination arm. Both trials showed increased toxicities with the combination chemotherapy regimens as compared to gemcitabine. The current second-line treatment option was established by the NAPOLI-1 trial, which compared a nanoliposomal irinotecan plus 5FU combination to single-agent nanoliposomal irinotecan or 5FU in patients who were previously treated with gemcitabine-based chemotherapy.14 Median OS was 6.1 months in the combination arm, compared to 4.2 months with single-agent 5FU.15 More recently pembrolizumab was approved for microsatellite instability-high (MSI-H) tumors in the second-line setting based on data from 149 patients across five single-arm clinical trials.16

Other cytotoxic chemotherapy agents are being explored in clinical trials. The phase I/II NAPOX trial combined nanoliposomal irinotecan, 5FU/leucovorin, and oxaliplatin in the first-line setting, and reported promising anti-tumor activity – six partial responses in 24 patients in one cohort.17 Clinical trials with cytotoxic chemotherapy combinations such as gemcitabine, capecitabine, cisplatin, irinotecan and trifluridine/tipiracil, and nanliposomal irinotecan are underway (ClinicalTrials.gov identifier: NCT03535727).18

Despite modest improvements in OS with cytotoxic chemotherapy, the 5-year OS of PDAC is still 8% and there is an urgent need for novel, effective systemic treatment approaches.

What systemic therapies are there on the horizon beyond cytotoxic chemotherapy?

Molecular characterization and genomic profiling of PDAC has led to a better understanding of the disease and currently there are more than 150 actively accruing clinical trials with novel agents exploring new treatment options and breakthroughs. These novel clinical trials focus on immune system, targeted therapy, pancreatic stroma, and tumor microenvironment. Promising targets for therapy in pancreatic cancer include driver oncogenes, DNA repair pathway, stem cell, and metabolomic pathway.

Emerging therapeutic targets for pancreatic cancer

Immunotherapy

Immunotherapy with anti-programmed cell death protein 1 (PD-1), anti-programmed death-ligand 1 (PD-L1), and anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibody treatment has become standard of care in several malignancies; however, the outcomes in PDAC with single agent or dual immune checkpoint inhibitors have been disappointing.19,20 The profoundly immunosuppressive tumor microenvironment of PDAC, dense stroma, inhibitory cytokines, low effector T-cell, and low mutational tumor burden have been implicated in the failure of immunotherapy in pancreatic cancer.21,22 Additionally, the PDAC tumor microenvironment is infiltrated with immunosuppressor elements, such as myeloid-derived suppressor cells, tumor-associated macrophages (TAMs), and neutrophils. Patients with PDAC and high CD8 T-cell infiltration or neoantigen numbers have been shown to have longer survival.23 Presence of higher effector T cells and lower alternatively activated macrophages (M2) are also associated with longer survival.24,25 Other than in subgroups of patients with PDAC with MSI-H tumors, use of checkpoint inhibitors in PDAC is still investigational.

Table 1

Microsatellite instability-high subgroup

Approximately 1% of patients with PDAC have MSI-H-positive disease.26 The immune checkpoint inhibitor pembrolizumab has shown a 53% objective response rate (ORR) across 12 solid tumor types, including pancreatic cancer.27 Pembrolizumab is currently approved for this indication in the second-line setting.27 Currently, a phase I study with anti-PD-L1 antibody LY3300054, alone or in combination with other agents, is enrolling patients with treatment refractory MSI-H solid tumors, including pancreatic cancer (ClinicalTrials.gov identifier: NCT02791334).

Immunotherapy and stromal targeting agents

Two main approaches to enhance immune therapy effects in PDAC, are being evaluated. The first approach is aimed at enhancing the activity of the immune checkpoint blockade by adding agents that address the immune inhibitory environment. These include rational combination approaches with immunomodulatory chemotherapy agents, radiotherapy, and small molecular agents, focusing on the tumor microenvironment for a more immune-favorable state, improving antigen presentation, and decreasing regulatory T-cells (Table 1). In the second approach, investigators are evaluating immune therapy approaches that do not include checkpoint blockade.

Checkpoint inhibitor-based combination approaches

Chemotherapeutic agents such as gemcitabine and cyclophosphamide have been shown to decrease regulatory T cells,28 which prompted early-phase clinical trials in combination with checkpoint inhibitors (Table 1). A combination of Gemcitabine plus tremelimumab was studied in a phase Ib trial in metastatic PDAC, and two partial responses were reported.29

DNA damage is the main mechanism of cancer cell death by radiotherapy; however, more recently, the immunomodulatory effects of radiotherapy have been recognized.30 Radiation induces the release of proinflammatory mediators and damage-associated molecules such as high mobility group box 1 (HMGB1), heat shock proteins, ATP, and calreticulin;31 upregulates major histocompatibility complex class I expression, dendritic cell activation, immune checkpoint expression; and increases antigen presentation, cytotoxic T-cell recognition of irradiated cells, immunomodulatory cytokines, and inflammatory mediators.32,33 The synergistic effects of checkpoint-inhibitor and radiotherapy combination have been demonstrated in preclinical studies, and combination approaches are being explored in ongoing clinical trials (Table 1).34

TAMs have immunosuppressive and tumor-promoting features, and therefore have significant implications in carcinogenesis and progression of metastatic disease.35 TAMs and other myeloid cells secrete proangiogenic growth factors and immunosuppressive cytokines36 contributing to the immunosuppressive tumor microenvironment.35 TAMs can either have a proinflammatory (M1) or anti-inflammatory (M2) phenotype. An anti-inflammatory TAM M2 phenotype has immunosuppressive features and is associated with more aggressive tumors, increased angiogenesis, invasion, metastasis, and negative prognosis.37–39 The development of anti-inflammatory TAM M2 depends on growth factors such as colony stimulating factor-1 (CSF-1), also known as macrophage CSF. CSF-1 is activated by CSF-1 receptor (CSF-1R) and has crucial role in macrophage differentiation and survival.40 CSF-1R is expressed on myeloid-derived suppressor cells, neutrophils, and dendritic cells in the tumor microenvironment. CSF1-R mediated signals predominantly control the tumor promoting features of TAM; therefore, targeting tumor promoting CSF-1R is a viable anti-tumor strategy. Several clinical trials with CSF-R1 inhibitors in combination with checkpoint inhibitors, chemotherapy, and other immunotherapy agents are actively accruing patients (Table 1). Chemokines mediate immune-cell trafficking between bone marrow, peripheral tissues, inflammatory sites, and the tumor microenvironment.41 CCR2 and CCR5 are chemokine receptors that are expressed on myeloid cells in the tumor microenvironment and promote an immunosuppressive tumor microenvironment by recruiting inflammatory monocytes from bone marrow.42 CCR5 is also expressed on regulatory T cells and mediates their migration to the tumor microenvironment and promotes TAM M2 phenotype. Patients with PDAC with high C-C chemokine ligand (CCL)2 and low CD8 + T-cell infiltration have shorter survival compared to low CCL2 and high CD8 + T cells.43 Targeting the C-C chemokine receptor type (CCR)-2 axis with a CCR2 inhibitor has been shown to increase effector T cells, decrease regulatory T cells, and decrease tumor growth and metastasis in murine pancreatic cancer models.43 Anti-tumor effect was further increased when CCR inhibition was combined with gemcitabine in the same murine model. Based on this preclinical study, the oral CCR2 inhibitor PF-04136309 in combination with FOLFIRINOX or FOLFIRINOX alone was studied in a phase Ib trial in borderline resectable and locally advanced PDAC.43,44 A partial response rate of 48.5% was reported with the combination therapy and treatment was tolerable.44 Investigation into a CCR2/CCR5 dual inhibitor in combination with gemcitabine, nab-paclitaxel, and nivolumab in borderline resecable or locally advanced PDAC is ongoing (ClinicalTrials.gov identifier: NCT03496662). A trial with CCR2/CCR5 dual antagonist in combination with chemotherapy or nivolumab is also ongoing in metastatic pancreatic and colorectal cancer.42

table 2

Tumor microenvironment and stroma

Dense stromal desmoplastic reaction is one of the hallmarks of pancreatic cancer tumor microenvironment.45 The fibrous tissue which constitutes the desmoplastic reaction is produced by pancreatic satellite cells. Pancreatic satellite cells also produce extracellular matrix proteins, cytokines, and vascular endothelial cells.21,46 The stromal desmoplasia is evident in both primary pancreatic tumors and metastatic tumor sites and creates a more hypoxic tumor microenvironment.47 In PDAC, the stroma is highly fibrotic, which creates a hypoxic tumor microenvironment and is implicated in tumor progression, metastasis, and treatment resistance; although it can have tumor restraining effects.21,48 Novel strategies targeting the stroma and tumor microenvironment, involving hedgehog pathway and Bruton’s kinase, have resulted in disappointing outcomes.49,50 Current strategies are focusing on targeting hyaluronic acid (HA), vitamin D receptor, and CD40 antibody (see Table 2).

HA is abundant in pancreatic stoma; it increases interstitial pressure, impacts oncogenesis, and is involved in cellular signaling pathway by binding to cell surface receptors, such as CD44.51–53 Pegvorhyaluronidase alfa (PEGPH20), a pegylated version of recombinant human hyaluronidase, has been shown to decrease HA enzymatically, and has reestablished and improved vascular permeability and improved intratumoral drug delivery in a preclinical study.54 PEGPH20 in combination with gemcitabine also resulted in higher survival and reduction in tumor size compared to gemcitabine alone in the same mouse model.54 The phase II HALO 202 trial compared PEGPH20 plus nab-paclitaxel/gemcitabine versus nab paclitaxel/gemcitabine regimen in untreated metastatic PDAC and reported improved progression-free survival (PFS), 6 months versus 5.3 months, respectively.55 When patients with HA-high tumors were analyzed, both the PFS and ORR was higher in the PEGPH20 plus nab-paclitaxel/gemcitabine arm, 9.2 months and 45% versus 5.2 months and 31%, respectively. Due to a higher rate of thromboembolic events in the PEGPH20 plus nab-paclitaxel/gemcitabine arm, this study was amended after the first stage, and enoxaparin prophylaxis was implemented in both arms. A clinical trial with PEGPH20 plus nab-paclitaxel/gemcitabine versus nab-paclitaxel/gemcitabine in HA-high PDAC is ongoing (ClinicalTrials.gov identifier: NCT02715804). Contrary to favorable outcomes with the GA combination, a phase I/II trial with PEGPH20 plus mFOLFIRINOX versus mFOLFIRINOX in untreated PDAC population was recently closed to accrual due to detrimental effect of OS.56

CD40 belongs to the tumor necrosis factor superfamily and is expressed by antigen-presenting cells as well as tumor cells.57 CD40 activation stimulates antigen presentation by the antigen-presenting cells, triggers proinflammatory cytokine release, enhances T-cell activation, and induces stromal depletion in both preclinical pancreatic cancer models and humans.57,58 The synergistic effects of CD40 agonist and cytotoxic chemotherapy combination has been shown in preclinical studies when CD40 was administered following gemcitabine.59 In another preclinical study with a KPC mouse model, CD40 activation redirected tumor infiltrating monocytes and TAMs and induced degradation of fibrosis, hence increased efficacy of chemotherapy.60 Interestingly, the sensitivity to chemotherapeutic agent was enhanced even when administered just days after CD40 antibody, indicating rapid degradation of fibrosis in the tumor model. The immunomodulatory effects of CD40 in the tumor microenvironment and antifibrotic properties in tumor stroma prompted further exploration of anti-CD40 monoclonal antibodies in clinical trials (Table 2). In a phase I study of 22 patients with advanced PDAC, the combination of agonistic CD40 monoclonal antibody with gemcitabine was well tolerated and achieved 19% ORR.61

PDAC stroma expresses vitamin D receptor; in preclinical study with a KPC tumor model, vitamin D analogue, calcipotriol, was shown to induce stromal remodeling, increase intratumoral vasculature, improve intratumoral drug delivery, and increase anti-tumor response when combined with gemcitabine.62 A phase II study combining with gemcitabine, cisplatin, nab-paclitaxel, paricalcitol, and nivolumab is currently enrolling; preliminary results from 10 patients in the initial phase of the study have revealed 80% ORR.63 A first-line placebo-controlled phase II clinical trial with the vitamin D agonist, paricalcitol, plus gemcitabine/nab-paclitaxel is currently accruing (ClinicalTrial.gov identifier: NCT03520790). Pilot studies are being conducted in resectable pancreatic cancer with vitamin D analogs and chemotherapeutic agents (ClinicalTrial.gov identifier: NCT02030860) and with checkpoint inhibitors (ClinicalTrial.gov identifier: NCT02930902).

Targeting DNA damage response and other pathways

Pancreatic adenocarcinoma has a complex genomic landscape and further molecular characterization has led identification of unique subgroups with different mutational features which may benefit from molecularly targeted therapy. Waddel et al. performed whole-genome sequencing of 100 patients with PDAC and demonstrated that chromosomal structure variation is a significant mechanism of DNA damage in PDAC.64 As such, PDAC has been classified into four different subtypes: stable subtype, locally rearranged subtype, scattered subtype, and unstable subtype. Scattered subtype has <200 structural abnormalities whereas unstable type has >200 structural abnormalities and mutations in DNA repair pathways such as BRCA1/2, PALB2, and ATM, which are more sensitive to platinum-containing regimens. A recent comprehensive genomic analysis of 456 pancreatic cancer cases revealed commonly mutated genes that aggregate into 10 cellular pathways.65 Based on gene expression, four different subtypes have been identified including squamous, pancreatic progenitor, immunogenic, and aberrantly differentiated endocrine exocrine. The gene expression features of these subtypes were correlated with histopathological characteristics and survival.

KRAS mutation is seen in 95% of PDACs and TP53, CDKN2A, SMAD4/DPC4, MLLSM, and RBM10 are other well-known mutations; however, the frequency of potentially actionable mutations with effective targeted therapies is much lower.66,67 Certain mutations such as NTRK gene fusions are highly actionable with very effective targeted therapy but NTRK gene fusions are detected in less than 1% of PDACs.68,69 Prominent investigational targeted therapy approaches include DNA repair pathways, cancer stem cells, metabolic pathways, and asparagine depletion.

DNA repair

Molecular subtyping of pancreatic cancer may have therapeutic implications as different subtypes may respond to therapy differently.70 Disrupted DNA damage response (DDR) is one of the hallmarks of pancreatic cancer development and is seen in up to 24% of PDACs,66 which also opens avenues for cancer treatment by utilizing synthetic lethality by targeting complementary DNA repair mechanisms by platinum agents and PARP inhibitors.71 BRCA1/2, PALB2, and ATM genes are key DNA maintenance genes in pancreatic cancer development.72 The majority of unstable type tumors have shown high BRCA mutational signature and/or unstable genome.64 BRCA-associated PDAC has superior survival with platinum-containing regimens and responds better to PARP inhibitors.73 Significant clinical activity with an ORR of 77.8% with PARP the inhibitor veliparib in combination with gemcitabine and cisplatin was reported in a phase I clinical trial in BRCA-mutated PDAC.74 Currently, a randomized phase II trial with and without veliparib combined with gemcitabine and cisplatin is enrolling patients (ClinicalTrials.gov identifier: NCT01585805) along with several other trials with PARP inhibitors (Table 3). The POLO trial randomized patients with BRCA-mutated metastatic PDAC to olaparib versus placebo (3:2 ratio) as a maintenance therapy after disease stability on a platinum containing regimen.75 The study has achieved its primary endpoint of improvement in PFS, which establishes an evidence-based approach for maintenance therapy in platinum sensitive BRCA-mutated PDAC. DDR mutations such as ATM, ATR, ATRX, BAP1, BARD1, BRIP1, CHEK 1/2, RAD50/51B, FANCA/C/D2/E/F/G/L beyond BRCA 1/2, and PALB2 are associated with improved OS when treated with platinum-containing regimens in advanced pancreatic cancer.76

Cancer stem cells

Cancer stem cells are highly tumorigenic, they have the ability to self-renew, can differentiate into heterogenous nontumorigenic cancer cell types comprising the tumor, are able to form spheres in stem cell media, and are resistant to traditional chemotherapeutic agents.77–80 A preclinical study with cancer stemness inhibitor BBI608, napabucasin, in a PaCa-2 pancreatic xenograft model, inhibited tumor growth, decreased stemness-high cancer cells in vivo, and down-regulated cancer stem cell proliferation and STAT3-driven self-renewal genes as compared to gemcitabine and carboplatin.78 In a phase Ib extension study with napabucasin and gemcitabine/nab-paclitaxel combination, no dose limited toxicity was encountered; in 29 evaluable patients, a 93% disease control rate and 79.3% ORR was reported in previously untreated PDAC.81 In a phase Ib/II study with the same combination, in 55 evaluable patients, a 93% disease control rate and 55% ORR was reported.82 Based on these encouraging results, a phase III trial with nababucasin plus gemcitabine/nab-paclitaxel is currently ongoing.83

Metabolic pathways

The mitochondrial metabolism inhibitor CPI-613 targets mitochondrial energy metabolism in cancer cells and induces apoptosis and autophagy.84 In a preclinical pancreatic cancer xenograft tumor model, significant anti-tumor activity was observed.84 A phase I trial with CPI-613 combined with mFOLFIRINOX reported 61% ORR, with hyperglycemia, hypokalemia, and peripheral neuropathy being the most common grade 3–4 nonhematological adverse events.85 CPI-613 is being explored in a phase III trial in combination with mFOLFIRINOX (ClinicalTrials.gov identifier: NCT03504423) and phase I trial with in combination with gemcitabine/nab-paclitaxel (ClinicalTrials.gov identifier: NCT03435289).

Erythrocyte-encapsulated L-asparaginase (eryaspase) has emerged as a potential therapeutic option for PDAC. Asparagine has an essential role in protein and nucleotide synthesis, it also regulates apoptosis, cell proliferation, and is essential for survival of pancreatic cancer cells.86–88 Asparagine synthase (ASNS) is induced by ATF4 and synthesizes asparagine from aspartate. Intracellular depletion of asparagine was shown to induce apoptosis.89 L-asparaginase depletes extracellular asparagine and it has anti-cancer effect on cancer cells with no or low ASNS expression.87 Interestingly, in lung cancer preclinical models, KRAS was identified as an important regulator in nutrient stress response in the cell, and through ATF4 regulation it was shown to alter asparagine biosynthesis, which is relevant in protein biosynthesis and apoptosis suppression.90 In the same preclinical study, ASNS was noted to be a key regulator of tumor cell proliferation. Disruption of the KRAS–ATF–ASNS pathway by inhibition of AKT sensitized tumor cells to L-asparaginase and was proposed as a potential treatment strategy in lung cancer. Eryaspase is encapsulated inside a donor-derived red blood cell, and anti-tumor activity by plasma asparagine depletion has been reported in a preclinical PDAC study.91 In a phase IIB study with chemotherapy +/- eryaspase, significant improvements in PFS and OS (26.1 for chemotherapy plus eryaspase versus 19.0 weeks for chemotherapy alone, p=0.03) were shown regardless of ASNS expression levels.92 An ongoing randomized phase III trial is exploring gemcitabine/nab-paclitaxel or an irinotecan-based regimen with or without eryaspase in second-line PDAC (ClinicalTrials.gov identifier: NCT03665441).

Conclusion

Clinical management of metastatic PDAC still depends on cyctotoxic chemotherapy. Although prevalence of MSI-H and NTRK gene fusions is low in PDAC, FDA-approved molecularly targeted therapy options are available for this subgroup of patients. Several promising therapies are in development based on strong scientific rationale including combination therapies with immune checkpoint inhibitors, stroma modifying agents, targeting of TAMs, DNA repair pathways, and cancer stem cells.

Article Information:
Disclosure

Mehmet Akce reports research funding from Merck Sharp & Dohme Corp., Eisai Inc., Xencor Inc., Tesaro, RedHill Biopharma, Polaris Pharmaceuticals, Bristol-Myers Squibb, and Ono Pharmaceutical, and has served in a consultatory/advisory role for Eisai Inc. Bassel F El-Rayes reports honoraria from Lexicon, RTI Health Solutions, and Bayer AG; has served in a consultatory/advisory role for Merrimack, BTG International, Bayer AG, Loxo Oncology, and RTI Health Solutions; is a member of the speakers’ bureau for Lexicon and Bristol-Myers Squibb; and declares research funding from Taiho Pharmaceutical (Inst), Bristol-Myers Squibb (Inst), Boston Biomedical (Inst), Cleave Biosciences (Inst), Genentech (Inst), AVEO Pharmaceuticals (Inst), Pfizer (Inst), Novartis (Inst), Hoosier Cancer Research Network (Inst), Five Prime Therapeutics (Inst), Pharmaceutical Product Development (Inst), Merck (Inst), and ICON (Inst).

Compliance With Ethics

This study involves a review of the literature and did not involve any studies with human or animal subjects performed by any of the authors.

Review Process

Double-blind peer review.

Authorship

The named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship of this manuscript, take responsibility for the integrity of the work as a whole, and have given final approval for the version to be published.

Correspondence

Bassel F El-Rayes, Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, 1365 Clifton Road NE, Suite C3003, Atlanta, GA 30322, USA. E: bassel.el-rayes@emoryhealthcare.org

Support

No funding was received in the publication of this article.

Received

18 March, 2019

References

  1. Rahib L, Smith BD, Aizenberg R, et al. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74:2913–21.
  2. Korc M, Jeon CY, Edderkaoui M, et al, Tobacco and alcohol as risk factors for pancreatic cancer. Best Pract Res Clin Gastroenterol. 2017;31:529–36.
  3. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.
  4. Gordon-Dseagu VL, Devesa SS, Goggins M, et al. Pancreatic cancer incidence trends: evidence from the Surveillance, Epidemiology and End Results (SEER) population-based data. Int J Epidemiol. 2018;47:427–39.
  5. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68:7–30.
  6. Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691–703.
  7. Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–25.
  8. Burris HA 3rd, Moore MJ, Andersen J, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15:2403–13.
  9. Berlin JD, Catalano P, Thomas JP, et al. Phase III study of gemcitabine in combination with fluorouracil versus gemcitabine alone in patients with advanced pancreatic carcinoma: Eastern Cooperative Oncology Group Trial E2297. J Clin Oncol. 2002;20:3270–5.
  10. Rocha Lima CM, Green MR, Rotche R, et al. Irinotecan plus gemcitabine results in no survival advantage compared with gemcitabine monotherapy in patients with locally advanced or metastatic pancreatic cancer despite increased tumor response rate. J Clin Oncol. 2004;22:3776–83.
  11. Louvet C, Labianca R, Hammel P, et al. Gemcitabine in combination with oxaliplatin compared with gemcitabine alone in locally advanced or metastatic pancreatic cancer: results of a GERCOR and GISCAD phase III trial. J Clin Oncol. 2005;23:3509–16.
  12. Oettle H, Richards D, Ramanathan RK, et al. A phase III trial of pemetrexed plus gemcitabine versus gemcitabine in patients with unresectable or metastatic pancreatic cancer. Ann Oncol. 2005;16:1639–45.
  13. Moore MJ, Goldstein D, Hamm J, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25:1960–6.
  14. Wang-Gillam A, Li CP, Bodoky G, et al. Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1): a global, randomised, open-label, phase 3 trial. Lancet. 2016;387:545–57.
  15. NCCN Clinical Practice Guidelines in Oncology. Pancreatic Adenocarcinoma. Version 1.2019-November 8, 2018. Available at: www.nccn.org/professionals/physician_gls/pdf/pancreatic.pdf (accessed February 2, 2019).
  16. Lemery S., Keegan P, Pazdur R. First FDA Approval Agnostic of Cancer Site — When a Biomarker Defines the Indication. N Eng J Med. 2017;377:1409–12.
  17. Dean AP, Wainberg ZA, Ramanathan RK, et al. A phase 1/2, open-label dose-escalation study of liposomal irinotecan (nal-IRI) plus 5- fluorouracil/leucovorin (5-FU/LV) and oxaliplatin (OX) in patients with previously untreated metastatic pancreatic cancer (mPAC). J Clin Oncol. 2018;36(15_suppl):4111.
  18. Alese OB, Shaib WL, Akce M et al. A phase I/II study of trifluridine/tipiracil (TAS-102) in combination with nanoliposomal irinotecan (NAL-IRI) in advanced GI cancers. J Clin Oncol. 2018;36(15_suppl):TPS4155.
  19. Royal RE, Levy C, Turner K, et al. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother. 2010;33:828–33.
  20. Brahmer JR, Drake CG, Wollner I, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28:3167–75.
  21. Young K, Hughes DJ, Cunningham D, Starling N. Immunotherapy and pancreatic cancer: unique challenges and potential opportunities. Ther Adv Med Oncol. 2018;10:1758835918816281.
  22. Lawrence MS, Stojanov P, Polak P, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013;499:214–8.
  23. Balachandran VP, Łuksza M, Zhao JN, et al. Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer. Nature. 2017;551:512–6.
  24. Ino Y, Yamazaki-Itoh R, Shimada K, et al. Immune cell infiltration as an indicator of the immune microenvironment of pancreatic cancer. Br J Cancer. 2013;108:914–23.
  25. Wang WQ, Liu L, Xu HX, et al. Infiltrating immune cells and gene mutations in pancreatic ductal adenocarcinoma. Br J Surg. 2016;103:1189–99.
  26. Hu ZI, Shia J, Stadler ZK, et al. Evaluating mismatch repair deficiency in pancreatic adenocarcinoma: challenges and recommendations. Clin Cancer Res. 2018;24:1326–36.
  27. Le DT, Durham JN, Smith KN, et al, Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–13.
  28. Cook AM, Lesterhuis WJ, Nowak AK, Lake RA. Chemotherapy and immunotherapy: mapping the road ahead. Curr Opin Immunol. 2016;39:23–9.
  29. Aglietta M, Barone C, Sawyer MB, et al. A phase I dose escalation trial of tremelimumab (CP-675,206) in combination with gemcitabine in chemotherapy-naive patients with metastatic pancreatic cancer. Ann Oncol. 2014;25:1750–5.
  30. Sharabi AB, Lim M, DeWeese TL, Drake CG, et al. Radiation and checkpoint blockade immunotherapy: radiosensitisation and potential mechanisms of synergy. Lancet Oncol. 2015;16:e498–509.
  31. Gameiro SR, Jammeh ML, Wattenberg MM, et al. Radiation-induced immunogenic modulation of tumor enhances antigen processing and calreticulin exposure, resulting in enhanced T-cell killing. Oncotarget. 2014;5:403–16.
  32. Reits EA, Hodge JW, Herberts CA, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med. 2006;203:1259–71.
  33. Finkelstein SE, Timmerman R, McBride WH, et al. The confluence of stereotactic ablative radiotherapy and tumor immunology. Clin Dev Immunol. 2011;2011:439752.
  34. Azad A, Yin Lim S, D’Costa Z, et al. PD-L1 blockade enhances response of pancreatic ductal adenocarcinoma to radiotherapy. EMBO Mol Med. 2017;9:167–80.
  35. Cannarile MA, Weisser M, Jacob W, et al. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J Immunother Cancer. 2017;5:53.
  36. Ries CH, Cannarile MA, Hoves S, et al. Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell. 2014;25:846–59.
  37. Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014;41:49–61.
  38. Ostuni R, Kratochvill F, Murray PJ, Natoli G. Macrophages and cancer: from mechanisms to therapeutic implications. Trends Immunol. 2015;36:229–39.
  39. Zhang QW, Liu L, Gong CY, et al. Prognostic significance of tumor-associated macrophages in solid tumor: a meta-analysis of the literature. PLoS One. 2012;7:e50946.
  40. Stanley ER, Chitu V. CSF-1 receptor signaling in myeloid cells. Cold Spring Harb Perspect Biol. 2014;6: pii: a021857.
  41. Griffith JW, Sokol CL, Luster AD. Chemokines and chemokine receptors: positioning cells for host defense and immunity. Annu Rev Immunol. 2014;32:659–702.
  42. Le D, Gutierrez ME, Saleh M, et al. Abstract CT124: A phase Ib/II study of BMS-813160, a CC chemokine receptor (CCR) 2/5 dual antagonist, in combination with chemotherapy or nivolumab in patients (pts) with advanced pancreatic or colorectal cancer. Cancer Research. 2018;78(13 Supplement):CT124.
  43. Sanford DE, Belt BA, Panni RZ, et al. Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: a role for targeting the CCL2/CCR2 axis. Clin Cancer Res. 2013;19:3404–15.
  44. Nywening TM, Wang-Gillam A, Sanford DE, et al. Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: a single-centre, open-label, dose-finding, non-randomised, phase 1b trial. Lancet Oncol. 2016;17:651–62.
  45. Nielsen MF, Mortensen MB, Detlefsen S. Key players in pancreatic cancer-stroma interaction: Cancer-associated fibroblasts, endothelial and inflammatory cells. World J Gastroenterol. 2016;22:2678–700.
  46. Haqq J, Howells LM, Garcea G, et al. Pancreatic stellate cells and pancreas cancer: current perspectives and future strategies. Eur J Cancer. 2014;50:2570–82.
  47. Whatcott CJ, Diep CH, Jiang P, et al. Desmoplasia in primary tumors and metastatic lesions of pancreatic cancer. Clin Cancer Res. 2015;21:3561–8.
  48. Neesse A, Algül H, Tuveson DA, Gress TM. Stromal biology and therapy in pancreatic cancer: a changing paradigm. Gut. 2015;64:1476–84.
  49. Catenacci DV, Junttila MR, Karrison T, et al. Randomized phase Ib/II study of gemcitabine plus placebo or vismodegib, a hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer. J Clin Oncol. 2015;33:4284–92.
  50. Kim EJ, Sahai V, Abel EV, et al. Pilot clinical trial of hedgehog pathway inhibitor GDC-0449 (vismodegib) in combination with gemcitabine in patients with metastatic pancreatic adenocarcinoma. Clin Cancer Res. 2014;20:5937–45.
  51. Provenzano PP, Hingorani SR. Hyaluronan, fluid pressure, and stromal resistance in pancreas cancer. Br J Cancer. 2013;108:1–8.
  52. Provenzano PP, Cuevas C, Chang AE, et al. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;21:418–29.
  53. Wong KM, Horton KJ, Coveler AL, et al. Targeting the tumor stroma: the biology and clinical development of pegylated recombinant human hyaluronidase (PEGPH20). Curr Oncol Rep. 2017;19:47.
  54. Jacobetz MA, Chan DS, Neesse A, et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut. 2013;62:112–20.
  55. Hingorani SR, Zheng L, Bullock AJ, et al. HALO 202: randomized phase II study of PEGPH20 plus nab-paclitaxel/gemcitabine versus nab-paclitaxel/gemcitabine in patients with untreated, metastatic pancreatic ductal adenocarcinoma. J Clin Oncol. 2018;36:359–66.
  56. Ramanathan RK, McDonough S, Agop Philip P, et al. A phase IB/II randomized study of mFOLFIRINOX (mFFOX) + pegylated recombinant human hyaluronidase (PEGPH20) versus mFFOX alone in patients with good performance status metastatic pancreatic adenocarcinoma (mPC): SWOG S1313 (NCT #01959139). J Clin Oncol. 2018;36(4_suppl):208.
  57. Vonderheide RH, Bajor DL, Winograd R, et al, CD40 immunotherapy for pancreatic cancer. Cancer Immunol Immunother. 2013;62:949–54.
  58. Beatty GL, Chiorean EG, Fishman MP, et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science. 2011;331:1612–6.
  59. Nowak AK, Robinson BW, Lake RA, Synergy between chemotherapy and immunotherapy in the treatment of established murine solid tumors. Cancer Res. 2003;63:4490–6.
  60. Long KB, Gladney WL, Tooker GM, et al. IFNgamma and CCL2 cooperate to redirect tumor-infiltrating monocytes to degrade fibrosis and enhance chemotherapy efficacy in pancreatic carcinoma. Cancer Discov. 2016;6:400–13.
  61. Beatty GL, Torigian DA, Chiorean EG, et al. A phase I study of an agonist CD40 monoclonal antibody (CP-870,893) in combination with gemcitabine in patients with advanced pancreatic ductal adenocarcinoma. Clin Cancer Res. 2013;19:6286–95.
  62. Kochenderfer JN, Somerville RPT, Lu T, et al. Lymphoma remissions caused by anti-CD19 chimeric antigen receptor T cells are associated with high serum interleukin-15 levels. J Clin Oncol. 2017;35:1803–13.
  63. Borazanci EH, Jameson GS, Borad MJ, et al. A phase II pilot trial of nivolumab (N) + albumin bound paclitaxel (AP) + paricalcitol (P) + cisplatin (C) + gemcitabine (G) (NAPPCG) in patients with previously untreated metastatic pancreatic ductal adenocarcinoma (PDAC). J Clin Oncol. 2018;36(4_suppl):358.
  64. Waddell N, Pajic M, Patch AM, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. 2015; 51:495–501.
  65. Bailey P, Chang DK, Nones K, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531:47–52.
  66. Dreyer SB, Chang DK, Bailey P, Biankin AV. Pancreatic cancer genomes: implications for clinical management and therapeutic development. Clin Cancer Res. 2017;23:1638–46.
  67. Borazanci E, Dang CV, Robey RW, et al. Pancreatic cancer: “a riddle wrapped in a mystery inside an enigma”. Clin Cancer Res. 2017;23:1629–37.
  68. Pishvaian MJ, Bender RJ, Halverson D, et al. Molecular profiling of patients with pancreatic cancer: initial results from the know your tumor initiative. Clin Cancer Res. 2018;24:5018–27.
  69. Pishvaian MJ, Garrido-Laguna I, Liu SV, et al. Entrectinib in TRK and ROS1 fusion-positive metastatic pancreatic cancer. JCO Precision Oncology. 2018; DOI: 10.1200/PO.18.00039.
  70. Collisson EA, Sadanandam A, Olson P, et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat Med. 2011;17:500–3.
  71. Perkhofer L, Illing A, Gout J, et al. Precision medicine meets the DNA damage response in pancreatic cancer. Oncoscience. 2018;5:6–8.
  72. Sahin IH, Lowery MA, Stadler ZK, et al. Genomic instability in pancreatic adenocarcinoma: a new step towards precision medicine and novel therapeutic approaches. Expert Rev Gastroenterol Hepatol. 2016;10:893–905.
  73. Golan T, Kanji ZS, Epelbaum R, et al. Overall survival and clinical characteristics of pancreatic cancer in BRCA mutation carriers. Br J Cancer. 2014;111:1132–8.
  74. O’Reilly EM, Lee JW, Lowery MA, et al. Phase 1 trial evaluating cisplatin, gemcitabine, and veliparib in 2 patient cohorts: Germline BRCA mutation carriers and wild-type BRCA pancreatic ductal adenocarcinoma. Cancer. 2018;124:1374–82.
  75. Golan T, Oh D-Y, Reni M, et al. POLO: A randomized phase III trial of olaparib maintenance monotherapy in patients (pts) with metastatic pancreatic cancer (mPC) who have a germline BRCA1/2 mutation (gBRCAm). J Clin Oncol. 2017;34(15_suppl):TPS4152.
  76. Pishvaian MJ, Blais EM, Brody JR, et al. Outcomes in pancreatic adenocarcinoma (PDA) patients (pts) with genetic alterations in DNA damage repair (DDR) pathways: Results from the Know Your Tumor (KYT) program. J Clin Oncol. 2019;37(4_suppl):191.
  77. Visvader JE, Lindeman GJ. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer. 2008;8:755–68.
  78. Li Y, Rogoff HA, Keates S, et al. Suppression of cancer relapse and metastasis by inhibiting cancer stemness. Proc Natl Acad Sci U S A. 2015;112:1839–44.
  79. Chen SF, Chang YC, Nieh S, et al. Nonadhesive culture system as a model of rapid sphere formation with cancer stem cell properties. PLoS One. 2012;7:e31864.
  80. Clarke MF, Dick JE, Dirks PB, et al. Cancer stem cells--perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res. 2006;66:9339–44.
  81. El-Rayes BF, Shahda S, Starodub A, et al. A phase Ib extension study of cancer stemness inhibitor BB608 (napabucasin) in combination with gemcitabine and nab-paclitaxel (nab-PTX) in patients (pts) with metastatic pancreatic cancer. J Clin Oncol. 2016;34(15_suppl):4128.
  82. Bekaii-Saab TS, Starodub A, El-Rayes BF, et al. A phase Ib/II study of cancer stemness inhibitor napabucasin (BBI-608) in combination with gemcitabine (gem) and nab-paclitaxel (nabPTX) in metastatic pancreatic adenocarcinoma (mPDAC) patients (pts). J Clin Oncol. 2017;35(15_suppl):4106.
  83. Bekaii-Saab TS, Li C-P, Okusaka T, et al. CanStem111P trial: A phase III study of napabucasin (BBI-608) plus nab-paclitaxel (nab-PTX) with gemcitabine (gem) in adult patients with metastatic pancreatic adenocarcinoma (mPDAC). J Clin Oncol. 2017;35(15_suppl):TPS4148.
  84. Zachar Z, Marecek J, Maturo C, et al. Non-redox-active lipoate derivates disrupt cancer cell mitochondrial metabolism and are potent anticancer agents in vivo. J Mol Med (Berl). 2011;89:1137–48.
  85. Alistar A, Morris BB, Desnoyer R, et al. Safety and tolerability of the first-in-class agent CPI-613 in combination with modified FOLFIRINOX in patients with metastatic pancreatic cancer: a single-centre, open-label, dose-escalation, phase 1 trial. Lancet Oncol. 2017;18:770–8.
  86. Fung MKL, Chan GC. Drug-induced amino acid deprivation as strategy for cancer therapy. J Hematol Oncol. 2017;10:144.
  87. Yee NS. Update in systemic and targeted therapies in gastrointestinal oncology. Biomedicines. 2018;6: pii: E34.
  88. Krall AS, Xu S, Graeber TG, et al. Asparagine promotes cancer cell proliferation through use as an amino acid exchange factor. Nat Commun. 2016;7:11457.
  89. Zhang J, Fan J, Venneti S, et al. Asparagine plays a critical role in regulating cellular adaptation to glutamine depletion. Mol Cell. 2014;56:205–18.
  90. Gwinn DM, Lee AG, Briones-Martin-Del-Campo M, et al. Oncogenic KRAS regulates amino acid homeostasis and asparagine biosynthesis via ATF4 and alters sensitivity to L-asparaginase. Cancer Cell. 2018;33:91–107.
  91. Dufour E, Gay F, Aguera K, et al. Pancreatic tumor sensitivity to plasma L-asparagine starvation. Pancreas. 2012;41:940–8.
  92. Hammel P, Bachet J-B, Portales F, et al. A phase 2b of eryaspase in combination with gemcitabine or FOLFOX as second-line therapy in patients with metastatic pancre­atic adenocarcinoma (NCT02195180). Ann Oncol. 2017;28(suppl_5):mdx369.005.

Further Resources

Share this Article
Related Content In Pancreatic Cancer
  • Copied to clipboard!
    accredited arrow-down-editablearrow-downarrow_leftarrow-right-bluearrow-right-dark-bluearrow-right-greenarrow-right-greyarrow-right-orangearrow-right-whitearrow-right-bluearrow-up-orangeavatarcalendarchevron-down consultant-pathologist-nurseconsultant-pathologistcrosscrossdownloademailexclaimationfeedbackfiltergraph-arrowinterviewslinkmdt_iconmenumore_dots nurse-consultantpadlock patient-advocate-pathologistpatient-consultantpatientperson pharmacist-nurseplay_buttonplay-colour-tmcplay-colourAsset 1podcastprinter scenerysearch share single-doctor social_facebooksocial_googleplussocial_instagramsocial_linkedin_altsocial_linkedin_altsocial_pinterestlogo-twitter-glyph-32social_youtubeshape-star (1)tick-bluetick-orangetick-red tick-whiteticktimetranscriptup-arrowwebinar Sponsored Department Location NEW TMM Corporate Services Icons-07NEW TMM Corporate Services Icons-08NEW TMM Corporate Services Icons-09NEW TMM Corporate Services Icons-10NEW TMM Corporate Services Icons-11NEW TMM Corporate Services Icons-12Salary £ TMM-Corp-Site-Icons-01TMM-Corp-Site-Icons-02TMM-Corp-Site-Icons-03TMM-Corp-Site-Icons-04TMM-Corp-Site-Icons-05TMM-Corp-Site-Icons-06TMM-Corp-Site-Icons-07TMM-Corp-Site-Icons-08TMM-Corp-Site-Icons-09TMM-Corp-Site-Icons-10TMM-Corp-Site-Icons-11TMM-Corp-Site-Icons-12TMM-Corp-Site-Icons-13TMM-Corp-Site-Icons-14TMM-Corp-Site-Icons-15TMM-Corp-Site-Icons-16TMM-Corp-Site-Icons-17TMM-Corp-Site-Icons-18TMM-Corp-Site-Icons-19TMM-Corp-Site-Icons-20TMM-Corp-Site-Icons-21TMM-Corp-Site-Icons-22TMM-Corp-Site-Icons-23TMM-Corp-Site-Icons-24TMM-Corp-Site-Icons-25TMM-Corp-Site-Icons-26TMM-Corp-Site-Icons-27TMM-Corp-Site-Icons-28TMM-Corp-Site-Icons-29TMM-Corp-Site-Icons-30TMM-Corp-Site-Icons-31TMM-Corp-Site-Icons-32TMM-Corp-Site-Icons-33TMM-Corp-Site-Icons-34TMM-Corp-Site-Icons-35TMM-Corp-Site-Icons-36TMM-Corp-Site-Icons-37TMM-Corp-Site-Icons-38TMM-Corp-Site-Icons-39TMM-Corp-Site-Icons-40TMM-Corp-Site-Icons-41TMM-Corp-Site-Icons-42TMM-Corp-Site-Icons-43TMM-Corp-Site-Icons-44TMM-Corp-Site-Icons-45TMM-Corp-Site-Icons-46TMM-Corp-Site-Icons-47TMM-Corp-Site-Icons-48TMM-Corp-Site-Icons-49TMM-Corp-Site-Icons-50TMM-Corp-Site-Icons-51TMM-Corp-Site-Icons-52TMM-Corp-Site-Icons-53TMM-Corp-Site-Icons-54TMM-Corp-Site-Icons-55TMM-Corp-Site-Icons-56TMM-Corp-Site-Icons-57TMM-Corp-Site-Icons-58TMM-Corp-Site-Icons-59TMM-Corp-Site-Icons-60TMM-Corp-Site-Icons-61TMM-Corp-Site-Icons-62TMM-Corp-Site-Icons-63TMM-Corp-Site-Icons-64TMM-Corp-Site-Icons-65TMM-Corp-Site-Icons-66TMM-Corp-Site-Icons-67TMM-Corp-Site-Icons-68TMM-Corp-Site-Icons-69TMM-Corp-Site-Icons-70TMM-Corp-Site-Icons-71TMM-Corp-Site-Icons-72