Submit manuscript...
Journal of
eISSN: 2373-633X

Cancer Prevention & Current Research

Review Article Volume 7 Issue 4

The molecular connections between lung and pancreatic cancer metastases: are we not seeing the forest for the trees?

Angelo Wilson

Southern Illinois University, USA

Correspondence: Angelo Wilson, Southern Illinois University, P.O. Box 92477, Washington, DC, USA, Tel 20018-2477

Received: November 04, 2016 | Published: February 22, 2017

Citation: Wilson A. The molecular connections between lung and pancreatic cancer metastases: are we not seeing the forest for the trees? J Cancer Prev Curr Res. 2017;7(4):111-126. DOI: 10.15406/jcpcr.2017.07.00242

Download PDF

Abstract

Metastases to the pancreas are rare at 1-2%. Among primaries metastasizing to the pancreas, lung cancer (LC) is frequently the culprit. Metastases to the lung from pancreatic cancer (PC) are significantly more common at about 45%, presenting striking differences in cancer behavior. There are conflicting reports regarding cancerogenicity and metastatic incidence between the lung and pancreas. Therefore, this review takes a fresh look at lung and pancreatic cancer behavior. Secondary metastases to the lung and pancreas are often indistinguishable from LC and PC primaries, and the seed and soil hypothesis is not always congruous with clinical interpretation of disease course. Sometimes single “seed” dissemination is thought to occur at the preneoplastic stage without any evidence of tumor invasion. Metastatic growth may become dormant, manifesting years later as cancer of unknown primary (CUP). Interestingly, CUPs are discovered to originate most frequently from LC and PC primaries at autopsy. The lung and pancreas are morphologically related through endoderm-level morphogenesis. The molecular basis of cancer regularity between them involves developmental signaling pathways including HEDGEHOG, NOTCH, WNT, and CXCL12/CXCR4, an evolving genetic background, and regulatory tumor-stromal interactions. Perhaps biomarkers that explain the regularity between them have been documented - as we peruse the bioscience literature for information, we may be reading through viable biomarkers and solutions and not seeing the forest for the trees. On the other hand, perhaps more research is warranted to explain cancer behavior between the lung and pancreas. Observations in this review provide a framework on which to extract clues for future work regarding organ-specific metastasis between the lung and pancreas.

Keywordscancer of unknown primary site, tumor dormancy, preneoplasia, seed and soil hypothesis, tumor heterogeneity, developmental signaling pathways,regulatory tumor-stromal interactions, genetic background, EMT/MET, pan-cancer analysis, diagnosed as secondary, best treatment option, molecular processes, difficult to distinguish, small cell

Abbreviations

LC, lung cancer; NSCLC, non-small cell lung cancer; LADC, lung adenocarcinoma; SCC, squamous cell carcinoma; LCNEC, large cell neuroendocrine carcinomas; Other Type, LCC, large cell carcinomas, other type; NSCLC–ASCC, adenosquamous cell carcinoma; SCLC, small cell lung cancer; BSD, bronchial squamous dysplasia; CIS, carcinoma in situ; AAH, atypical adenomatous hyperplasia; PNCH, pulmonary neuroendocrine cell hyperplasia; PC, pancreatic cancer; PDAC, pancreatic ductal adenocarcinoma; Panin, pancreatic intraepithelial neoplasia; PMCN, mucinous cystic neoplasms; IPMN, intraductal papillary mucinous neoplasm; PNET/ICT, pancreatic neuroendocrine tumors/islet cell tumors; PACC, pancreatic acinar cell carcinoma; CUP, cancer of unknown primary site; Hh, hedgehog; EMT, epithelial to mesenchymal transition; MET, mesenchymal to epithelial transition; CTC, circulating tumor cell; Cscs, cancer stem cells; HNC, head and neck cancer; Goqs, guiding observational questions

Introduction

Lung cancer (LC) remains the number one cause of cancer death in the United States (US) and the world, with an expectation of 222,500 new cases and 155,870 deaths in the US in 2017.1 About 80% of LC deaths in the US are caused by smoking, but trends are declining, as falling mortality rates reflect smoking reduction.2,3 LC is often diagnosed at advanced metastatic stage, and is expected to remain the leading cancer killer beyond the year 2030.1,3,4 Meanwhile, pancreatic cancer (PC) is expected to be the 3rd leading cause of cancer-related death in the US in 2017, and like LC, risk is significantly increased in smokers compared to never smokers.1,2 There will be 53,670 new PC cases and 43,090 deaths in the US in 2017.1 Most alarming is that PC is expected to become the second leading cause of cancer death by 2030, surpassing colorectal cancer.1,3 Although survival rates have improved for several cancers, the same is not true for PC. Overall survival (OS) remains dismal for PC patients treated by surgery, and like LC patients, they often succumb to metastatic disease.3,5,6 Surgery is the best treatment option in most cases, but most patients do not qualify for surgery. Without surgery, the OS rate is 3-6months.7 Surgery alone is not curbing mortality trends, and the median OS rate of resected patients receiving adjuvant chemotherapy and/or radiation therapy after surgery is only 20-23 months.8 Taken together, over half of all LC and PC cases are diagnosed at a distant stage, for which 5-year survival rates are a dismal 4% and 2%, respectively.2 These data suggest that molecular research, and randomized and blinded clinical trials that focus on prevention and early-stage detection, and advanced-stage molecular therapy, must be continued.9,10

Cancer behavior refers to variations in growth, malignant progression, and morbid systemic spread of tumors, through wide-ranging degrees of severity.11,12 The totality of biological properties that explain these events include but are not limited to: mutations, genetic background, signaling pathway redundancy, pathway interactions (epistasis), gene pleiotropy, tumor-stromal interactions (TSIs), epithelial-to-mesenchymal (EMT) processes, tumor dormancy, angiogenesis, and histological subtype.13,14 Cancer behavior can range on a continuum from benign/indolent to aggressively metastatic.15‒17 Biological mechanisms that explain variations in cancer behavior are not completely known.12

Molecular biology and (sometimes) organ specificity determine where a tumor spreads (if it spreads at all) and the ensuing severity of metastasis.12 Sometimes metastasis is undetectable, asymptomatic, and discovered incidentally-also known as "occult metastasis”-or it is discovered in recurrent assessments or autopsy.18 Sometimes metastasis is diagnosed as secondary cancer of unknown primary (CUP), also known as "occult cancer".19,20 Determination of the unknown primary site is often a critical challenge for clinicians. CUP is expected to be the 4th most common cause of cancer death in the U.S. in 2017, and although trends are decreasing, there are not many recent reports that specifically examine cancer biology leading up to CUP to reflect this trend, while diagnoses remain inconsistent, and prognoses mixed.1,21,22 At autopsy, CUPs are determined to originate from a primary LC or PC in most cases.23‒24 Sometimes CUPs develop from dormancy or growth regression of primaries.23,24 CUP patients often do not respond to adjuvant therapy, and cancer stem cells (CSCs) are believed responsible.24 As discussed in this review, CSCs are being researched as targets for therapy regarding proliferation, dissemination potential, and organ-specific metastasis.25

Complications due to metastatic disease and resistance to therapy are the common cause of death in LC and PC patients.26‒29 LC and PC primaries metastasize to organ-specific sites more frequently than other systemic locations.11,12,30,31 Interpatient heterogeneity shows that there is regularity in metastasis between the organs of these diseases. The most frequent sites of metastasis for primary LC are the adrenal glands, bone, and liver.12,32 Metastases to the pancreas are extremely rare for all primaries at 1-2% and usually metachronous.32 Surveys on this topic varied, but of all primary cancers that metastasize to the pancreas, LC and renal cell carcinoma were found among the most common (as references indicate, there are conflicting data in the literature regarding LC metastasis to the pancreas).27,31‒34 Small cell lung cancer (SCLC) is the subtype that frequently metastasizes to the pancreas, however, primary non-small cell lung cancer (NSCLC) subtypes also metastasize to this organ.33 NSCLCs are divided into five different subtypes, all of which are prone to disseminate disease: lung adenocarcinoma (LADC), squamous cell carcinoma (SCC), adenosquamous cell carcinoma (ASCC), large cell carcinoma (LCC), and large cell neuroendocrine carcinoma (LCNEC).35 The oncogenic drivers that cause migration of SCLCs and NSCLCs to the pancreas are not fully understood. Conversely, the aggressive behavior of PC appears to be related to either early dissemination, late diagnosis, or a combination of the two.6,15,36,37 PC dissemination generally shows up in the liver, but can escape the liver and migrate to the lung, which is a common metastatic site for PC.6,12,27 Pancreatic adenocarcinoma (PDAC), pancreatic acinar cell carcinoma (PACC), and pancreatic neuroendocrine tumor (PNET) subtypes are aggressive to varying degrees and capable of widespread dissemination, but PDAC is the subtype often presented in lung metastasis.38 Secondary PDACs in the lung are difficult to distinguish from primary LCs because localization occurs along the alveoli, features a mucinous-type epithelium, and mimics primary bronchioloalveolar carcinoma.27,38 Likewise, secondary lesions to the pancreas are difficult to distinguish from primary PC.34,39 For PC patients, lung metastases offer a survival advantage over other metastatic sites, but no molecular basis has been put forth that explains this phenomenon.39‒41 There are no reports that indicate that the reverse is true - that pancreatic metastases from the lung offer a survival advantage over other metastatic sites for LC patients. These data suggest that the molecular processes that drive metastasis between these organs may have something to do with recapitulations of developmental signaling, and the fact that the organs are related through endoderm-level morphogenesis, of which Sonic Hedgehog (Hh), NOTCH, WNT, and CXCR4 are essential players.6,42‒45

Early tumor cell dissemination is believed to involve processes taking affect in preneoplastic, benign/indolent subtypes, including bronchial squamous dysplasia (BSD) carcinoma in situ (CIS), atypical adenomatous hyperplasia (AAH), and pulmonary neuroendocrine cell hyperplasia (PNCH) in the lung, and pancreatic intraepithelial neoplasia (PanIN), mucinous cystic neoplasms (MCN), and intraductal papillary mucinous neoplasm (IPMN) in the pancreas.46‒48 These preneoplastic subtypes feature biological mechanisms that can drive forward metastasis before malignant invasion.49‒52 Tumor progression processes in preneoplasia alter, transform, and diversify phenotypes, but are not completely understood.27,53‒55 Pipinikas et al.,56 showed that in situ precursory cells that are clonally related can physically migrate over distances, suggesting that multifocal preinvasive lesions in the progression from BSD to SCC originate from a common clonal ancestor, which endorses the field cancerization hypothesis of lung tumorigenesis.56 Yachida et al.,57 showed that PCs contain heterogeneous mixtures of subclones and each subclone contains millions of cells. The subclones are genetically evolved from the parental, non-metastatic clone, and are capable of metastasis. However, no molecular drivers were unveiled.57 These data suggest that clonally-regulated stem and progenitor cells, and microenvironmental processes in preneoplasia and early neoplasia, are involved in dissemination and metastasis in LC and PC.23,24,58‒60 Perhaps preneoplastic dissemination signals, and TSIs in the secondary organ conduce selective growth in the new microenvironment, and these effects along with genetic background, infringe on tumor indolency, dormancy, and metastatic CUP cases.24,61,62 Molecular profiling of TSIs may offer the possibility of screening and early detection and lead to improved diagnostics23,62‒64

An understanding of what is going on similarly and differently between metastatic LC and PC may help explain variations in cancer biology.31,32,33,27,34 It should be noted that all cancers acquire a similar set of capabilities necessary to manifest malignant disease. These general hallmarks of cancer were depicted in the classic article by Hanahan and Weinberg.65 Although at advanced stage neoplasia has disseminated to various locations in most cases, metastases to the pancreas, and to the lung, are frequently originated from one to the other respective organ, which merits some investigative attention.6,31,32,27,34,28 What are the greater molecular details that explain how and why this happens66‒69 The guiding observational questions (GOQs) are:

  1. Is there robust regularity in molecular processes that specify metastasis between the lung and pancreas? Are the drivers consistent? What biomarkers can be tested?
  2. What mechanisms confer resistance to secondary pancreatic lesions, making metastasis rare at 1-2% in clinical cases, and up to 11% at autopsy34 How are the mechanisms perturbed to allow frequential metastasis from LC primaries, and, what drives the perturbation?
  3. Conversely, what biological processes in pancreatic primaries prompt metastatic spread to lung to a significantly higher degree at 45%, when, as just stated, the reciprocal is seen in rare cases? What are the drivers behind these processes?
  4. Altered processes in pancreatic preneoplasias mediate succession toward ductal, neuroendocrine, and acinar carcinomas, sometimes in extremely rare performance.70 Likewise, preneoplasias in the lung spur malignant invasion, and may prompt cancerization, sometimes in extremely rare performance.71‒74 Are the isolated molecular processions repeated upon dissemination to spur metastasis? Are we in fact looking at reiterations that drive recurrence and metastasis between the two organs8,11,67 Are these the same processes that give rise to CUPs11,24 Do these processes interplay with tumor dormancy, the dormancy-to-proliferation switch, regression of primaries, and CSC plasticity.16,75

These GOQs establish the basis for this review, and designate origination of disease as an essential premise. Hypothetically, there are undetermined tumorigenic processes - biomarkers - in action that explain contrasting LC and PC behavior.6 On one hand, molecular processes resist secondary pancreatic metastases at a low occurrence of 1-2%, yet exhibit frequent organ-specific metastasis from LC primaries30,31,33,27,34 On the other hand, molecular drivers promote sensitivity to lung metastases from pancreatic primaries at a much higher occurrence rate of about 45%.27 Determining the molecular signatures could clarify contradistinctions in LC and PC metastasis, and lead to biomarkers that guide the engineering of personalized drugs.

Molecular connections between Lc and Pc

Recapitulated developmental signaling implicated in cancer behavior: Based on interpatient heterogeneity, the molecular basis of cancer regularity must be considered for effective personalized therapeutics. Signaling pathways and TSIs are directed by genetic background (reviewed in the next section) to conduce subtype-specific and organ-specific lung and pancreatic neoplasia.76‒79 Evidence shows that developmental signaling pathways that function most prominently in embryogenesis are aberrantly activated in neoplasia, recapitulating processes that drive development.80 Not only are Hh, NOTCH, WNT, and CXCR4 involved in endoderm-level development, they are also active in preneoplastic signaling, CSC activation, locoregional and disseminated tumor progression, and metastasis.80‒85 There are inhibitors for these pathways, some are approved by U.S. Food and Drug Administration, and more are being tested.86 However, drug resistance frequently occurs in LC and PC cases, and there are no standout reports examining developmental signaling to explain subtype-specific and organ-specific metastatic behavior between the lung and pancreas. Described here are Hh, NOTCH, WNT, and CXCR4, and their roles in LC and PC biologic behavior.

Hh signaling is virtually inactive in adults except for tissue repair and maintenance functions. Aberrant reactivation of Hh signaling is long known to play a role in lung and pancreatic tumorigenesis throughout the continuum from preneoplasia to metastasis, in a subtype specific manner.27,87,88 Watkins et al.,87 reported that Hh signaling activates lung neuroendocrine progenitor cells in the same way it functions in the maintenance and progression of SCLC.87 Under conditions of cellular stress, such as through drug intervention, the Hh pathway helps maintain cellular survival, growth and invasiveness, and thwarts cell destruction by signaling for effectors that stimulate drug resistance. Lin and colleagues examined the functions of Hh signaling in LDAC cells with acquired resistance to epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs).88 The authors found that the Hh pathway drives cancer progression in the face of EGFR-TKI antagonism. Hh interacting protein (HHIP) is a glycoprotein that binds to Hh ligands, shutting down Hh signaling. This glycoprotein is ablated through hypermethylation in pancreatic neoplasms making Hh fully operational. Lin and colleagues confirmed that HHIP is also shutdown in LADC, and overexpression of HHIP thwarted LADC growth in mice. Thus, the investigators present evidence that Hh is implicated in driving EGFR-TKI resistance in LADCs.88 Hao et al.,89 also investigated oncogenic Hh signaling in PC stem cell marker expression, cellular proliferation, progressive EMT processes, and tumor cell invasion, and the pathway was shown to co-regulate these processes.89 Hh also heavily crosstalks with the TGFβ and KRAS pathways, major players involved in tumor progression, EMT and MET processes, and metastasis.69,75,90,89 Taken together, these data show that the Hh pathway influences cancer behavior in both LC and PC lesions.

The NOTCH pathway plays several important roles in cancer progression, and its most critical role is at the nexus between invasion and metastasis Ni et al.,91 At this juncture, the NOTCH pathway crosstalks heavily with the EGFR pathway, and its communication is essential for reactivation of dormant CSCs and disease recurrence.92 Exhibit in their review that NOTCH is activated in pancreatic CSCs that have survived conventional therapy Ni et al.,91 Furthermore, NOTCH mediates pancreatic tumor relapse, and re-ignites growth and metastasis, and it does this thorough Notch-1 upregulation, induction of NF-κβ and its downstream effectors, expression of microRNAs (miRs) that regulate CSCs (miR21, miR200b, miR200c and miR34), induction of the EMT phenotype, heterogenic clonal expansion, and malignant migration Ni et al.,91 NOTCH performs the same operations in lung CSCs - which serves as a biomarker that defines lung CSCs due to the strength of the NOTCH signaling profile. Using GFP reporter recombinants and sphere-forming assays, Hassan’s group showed that NOTCH functions in heterogenic self-renewal (clonal heterogeneity). Through inoculation in mice, they also showed that NOTCH influences resistance to chemotherapy, and decreased LADC overall survival.93 These data suggest that recapitulated signaling through NOTCH is a vital force in LC and PC growth behaviors.

Wnt genes are downstream targets of the Hh pathway. WNT augments NOTCH signaling in tumor progression, and crosstalk frequently occurs.94,95 Both WNT and NOTCH co-regulate CSC biology, making WNT an equipotent therapeutic target.96,97 Interestingly, both pathways are involved in the re-vitalization of dormant cells, and recent reports reveal that turning off WNT signaling is implicated in metastatic dormancy and immune system evasion.98‒100 Pollard’s review implicates that inhibition of WNT serves to activate dormant epithelial cancer cells.99 Malladi et al.,100 confirm Pollard’s review, showing that autocrine inhibition of WNT signaling is code for evading the immune system and activating dormant tumor cells, implicating WNT inhibition in metastatic spreading.100 These reports are counterintuitive to the report by Wang’s group, who showed that inactivation of WNT through CXCL12/CXCR4 inhibits PC progression.101 Further confoundment emerges through evidence that CXCL12/CXCR4’s activation of the Hh pathway, which turns on WNT, induces invasion and EMT modulation.102 These seeming conflicts in cancerogenicity must be clarified, and likewise, the interplay of WNT and NOTCH in tumor dormancy and metastasis also must be clarified. If by further investigation these seeming conflicts connect and are justified, then per Malladi et al.,103 and Pollard’s review, WNT’s dysregulation can only be part of the story, because the dormancy-to-proliferation switch and angiogenesis also play roles in driving metastasis.16,103,104 As recently reviewed in Bleau and colleagues, the stroma co-regulates CSC sustenance, re-programming of the extracellular matrix, and the angiogenic switch;105 therefore, (putative) WNT dysregulation, and regulation from the stroma, drive forward cancer progression. These findings warrant further investigation regarding the interrelationships between NOTCH signaling, WNT signaling, stromal co-regulation of CSCs, the dormancy-to-proliferation switch, and angiogenesis, which could possibly help explain aggressive cancer behavior and drug resistance.16,25,106

The CXCL12/CXCR4 pathway is critical for LC and PC progression, and plays a role in metastatic homing of CSCs to specific organs.107,108 Recent reports show that it operates in stem cell niches with Hh and NOTCH in the progression of other cancers.109,110 CXCR4 is a recognized marker for CSCs, and this pathway may control frequential dissemination of PC tumor cells to the lung, and LC tumor cells to the pancreas.111,112 Interplay between CXCL12/CXCR4, Hh, and NOTCH may be consistent in LC and PC progression, which provides clues to GOQ-1. Furthermore, the CXCL12/CXCR4 pathway interacts with the stroma to influence neoplastic extravasation and the metastatic cascade.113 It also co-regulates immunosuppressive mechanisms.114,115 Pathway ligands are as critical as the CXCR4 receptor, as they can transduce signals through receptors other than CXCR4, driving oncogenic mechanisms through several pathways.116 For example, CXCL12 can bind to CXCR4 or CXCR7 to induce similar oncogenic mechanisms. Likewise, macrophage migration inhibitory factor (MIF) is an alternative ligand that can bind both CXCR4 and CXCR7 in place of CXCL12.117 Strikingly, THE CXCL12/CXCR4 pathway has not been studied extensively in a subtype-specific manner in preneoplastic lung and pancreatic lesions, such as BSD, CIS, AAH, PNCH, MCN, and IPMN.118 Kure et al.,119 reported a comparative study of CSC markers in PanIN-1, PanIN-2, and PanIN-3 vs. PDAC.119 Using patient-derived paraffin-embedded tissue, the investigators exhibited that both CXCR4 and epithelial-specific antigen (ESA), two proven morphoregulatory CSC markers, increased expression in the PanIN-to-PDAC progression. ESA displayed significant increases beginning at PanIN-1, and CXCR4 displayed significant increases beginning at PanIN-2. All CSC markers analyzed in the report were expressed in all stages of the progression. This suggests that upregulated CXCR4 expression in preneoplastic PanIN is indicative of proliferation, onset of migration, and metastasis, as they indicate in PDAC cells, making CXCR4 and its peripheral effectors of the chemokine-receptor network attractive biomarkers.119

Targeting intratumoral pathways has not been enough to curb LC and PC disease outcomes despite outstanding research.25 For example, targeting NOTCH alone may not be enough to abrogate drug resistance, as it has been shown that drug inhibition of NOTCH may induce squamous epithelial malignancies.120 The WNT pathway is fickle, and may prove unreliable as an isolated target aimed at alone. Whether WNT is inhibited or activated cancer progression ensues.84,95,97‒100 For example, SOX9 regulates the WNT pathway. One of WNT’s downstream targets is osteopontin (OPN), but the FGF pathway can alternatively activate OPN. If SOX9 or the WNT pathway are ablated, OPN can still be expressed as a downstream target of the FGF pathway - an exhibition of functional redundancy. Fully knowing regulatory mechanisms regarding the WNT pathway is not enough to stop OPN expression. Similar examples can be cited for Hh and CXCR4 pathways. This suggests that genetic background directs differences in cancer behavior and treatment response (Table 1).

Lung Cancer

Pancreatic Cancer

Nsclc - Ladc (40%)

Pdac (Ductal, 90%)

Nsclc - Scc (25-30%)

Pnet/Ict (Endocrine, 4%)

Nsclc - Lcc (5-10%)

Pacc (Acinar, 2%)

Nsclc - Lcnec (1.5-3%)

Panin (Precursory)

Nsclc - Ascc (1-2%)

Mcn (Precursory)

Sclc (15%)

Ipmn (Precursory)

BSD (Precursory)

CIS (Precursory)

AAH (Precursory)

PNCH (Precursory)

Table 1 Subtype classifications of lung and pancreatic cancers reviewed in this report
NSCLC, non-small cell lung cancer; LADC, lung adenocarcinoma; SCC, squamous cell carcinoma ; LCNEC, large cell neuroendocrine carcinomas other type; LCC, large cell carcinomas, other type; ASCC, adenosquamous cell carcinoma; SCLC, small cell lung cancer; BSD, bronchial squamous dysplasia; CIS, carcinoma in situ; AAH, atypical adenomatous hyperplasia; PNCH, pulmonary neuroendocrine cell hyperplasia; PDAC, pancreatic ductal adenocarcinoma; PNET/ICT, pancreatic neuroendocrine tumors/islet cell tumors; PACC, pancreatic acinar cell carcinoma; PANIN, pancreatic intraepithelial neoplasia pancreatic; MCN, mucinous cystic neoplasms; IPMN, intraductal papillary mucinous neoplasm

Genetic background: redundancy, epistasis, and pleiotropy in cancer behavior: Genetic background regulates recapitulated developmental signaling, and for this reason, combination therapy (a drug cocktail approach) is being researched as a treatment modality.66,121,122 Genetic background refers to the genomic and epigenomic landscapes that code for the molecular biology and survival mechanisms of the cell.61,123 All cancers feature a mutator phenotype, and the genetic background evolves through genomic instability and gradual widespread mutations.124,125 In sporadic cancer cases, genomic instability is often acquired by risk factors such as smoking.126,127 In heritable cases, genomic instability is prompted by inborn mutations, similar to acquired mutations in sporadic cases. The difference is that the molecular basis of heritable carcinogenesis is associated with predisposition and probability by family history, with disease being presented earlier, which further affirms genetic background as the producer of cancerogenicity.20,128‒130

There are cases where cancer was diagnosed as sporadic, although there was a heritable predisposition featured.131 Such cases relate to Dr. Alfred Knudson’s two-hit hypothesis.132 In heritable cases, the two-hit hypothesis refers to loss of heterozygosity (LOH); inactivation of both alleles of an oncogene (OG) or tumor suppressor gene (TSG) through inherited germline mutation(s) in one allele as the "first hit", and subsequent mutation(s) in the second allele (owing to genomic instability) as a "second hit”. In sporadic cases, both hits are acquired on an OG or TSG as somatic mutations.132 Germline haploinsufficiency in the background of somatic mutation(s) can influence sporadic oncogenesis.133,134 Recalling GOQ-4, it is quite possible that haploinsufficiencies influence sporadic CUP, rare cancer, and metastatic-like cancer cases, such as reported by Newman and colleagues, and Dr. Killen, respectively.11,70,73 Haploinsufficiencies among miR genes could predispose family members to heritable cancer, and influence oncogenesis and metastasis in sporadic cancer.135 These examples are digressions from Knudson’s two-hit hypothesis. Investigations that examine genetic background effects on heritable and sporadic cancer are warranted to generate specialized therapeutic approaches.

Genetic background effects also relate to the seed and soil hypothesis formulated by Dr. Stephen Paget.136 The seed and soil hypothesis refers to tumor cells that exhibit patterned migration and metastatic colonization in site-specific organs through premetastatic niche preparation.136 Lung metastasis is a common secondary incidence for known primaries, and this was thought to occur through cancer cells being trapped in pulmonary microvessels. Evidence suggests that a premetastatic niche is formed before arrival of lung CSCs, but how premetastatic niches are formed is still being investigated.137 Recent evidence suggests that exosomal content dispersion via tumor-released exosomes into blood circulation is implicated in preparing the premetastatic niche and immunosuppression.138 While the seed and soil hypothesis is robust in conceptualizing organ-specific metastasis, and is much appreciated regarding Dr. Paget’s intellectual insights, it is still hypothetical and subject to inspection. Metastasis in general is not completely understood.139 Disseminated tumor cells can become attached through cell surface adherence or microvessel size restrictions.139 Adhesive components on tumor cells (e.g., EpCAMs, integrins, glycolipids, etc.) can adhere to microvessel endothelial cells, subendothelial basement membrane matrix, and parenchymal cells.30,28,139,140 That being said, premetastatic niche is thought to not be causal in all metastatic cases.140,141 Recalling GOQ-2 and GOQ-3, genetic background likely plays a significant role in organ-specific LC and PC metastasis. If susceptibility to cancer incidence is increased through pathogenic germline mutations in lung and pancreatic cancer cells (seeds), then it seems plausible that stromal gene expression subsequently evolves in the invaded microenvironment (soil) to meet the survival needs of predisposed cells, especially where those cells are capable of adhering.142 It only takes one precancerous (founder) cell to initiate cancer, recurrence, or metastasis.143 In contrast to the seed and soil hypothesis, site-selective adhesion and genetic background might partly explain germline and sporadic rare metastases, including those presented in the pancreas.144,145 Recalling GOQ-1, pre-programmed CSCs may succeed through premetastatic niche or site-selective adhesion mechanisms, suggesting that the molecular processes in LC and PC metastasis are probably variable and inconsistent, and relevant biomarkers transiently change (as thought to happen in drug resistance), although interpatient regularity is clinically observed.12,28,110,114,143 Randomness with outcome regularity is evident through the recent findings by Martinez and colleagues that intratumor heterogeneity mimics intertumor heterogeneity.146 Investigations are needed to differentiate site-selective adhesion and premetastatic niche processes in metastasis. Moreover, rigorous testing of the two-hit and seed and soil hypotheses are warranted to further elucidate the genetic background impact on oncogenesis, and better potentiate individualized therapy approaches.81,147,148

Genetic background also impacts clonal diversity through parallel progression.11,28,125,149 The parallel progression model affirms that cancer behavior is largely due to genetic background; it is the accepted model to track cancer cells of origin. Evidence from this model serves as a paradigm shift from the former serial progression model.149 Observed features that endorse parallel progression are depicted in CUPs, variations in latency, and cases of metastasis of metastasis (intermetastatic heterogeneity).28,146 Although reports present evidence, pathway involvement in clonal origination is difficult to trace.150,151 It is not known whether a founder host stem cell, or induced cancerous cell that acquires stemness, originates oncogenesis.27,104,121,143 This is important because mutations produce detectable tumors on a time scale ranging from months to dozens of years, depending on background influences on tumor-stromal interplay.27,75,99,100,125,152,153 On one hand, metastatic CSCs originate from mutations in normal host stem cells through a process referred to as “pretumor progression”.150 On the other hand, invasive cancerous cells acquire a stem-like phenotype and then originate metastasis.27,104,143,150 Research must determine what driver mutations in cells of origin contribute to cancer cell fate and behavior.24,25,154,76,123,154 Validating background effects on cancerogenicity through redundant, interactive, and pleiotropic connections across the tumor-stromal axis could lead to effective therapeutic approaches.153,154

Genetic redundancy refers to paralogous genes that produce the same phenotype.155 Paralogous genes that functionally compensate one another implies their importance to cell survival, and fortifies Darwinian selection by reproductive fitness.155,156 Genetic redundancy does not necessarily imply functional redundancy, as paralogous gene products may have divergent functions elsewhere in the cell (e.g., moonlighting proteins).156‒158 The crosstalks of Hh, NOTCH, WNT and CXCR4 functionally succeed in proliferation of cancer phenotypes and variations thereof.80,159 Indeed, this is what we see in acquired and intrinsic drug resistance. Despite treatment with pathway inhibitors, LC and PC phenotypes and variations thereof continue to progress and metastasize.80,160 The ‘synthetic lethality’ approach aims to thwart oncogene addiction and pan-resistance - fortified disease progression through the exploits of functional redundancy.157,160 For example, CXCR4, NOTCH, and WNT pathways can turn on Hh downstream targets. If Hh is ablated, its downstream targets can still be expressed through crosstalks.147 Likewise, the Hh pathway crosstalks with other pathways such as TGFβ and IGF-R. If TGFβ and IGF-R pathways are abrogated, their downstream targets are still activated by Hh.160,161 Thus, genetic redundancy fortifies natural selection for cell survival (drug resistance).162 Combination therapy and synthetic lethality strategies are currently being explored to transform cancer phenotypes to lethal (apoptotic) phenotypes.66,157,160,162 Genetic background assessments on a case-by-case basis may improve clinical efficacy in such pursuits. The impact of epistasis regarding combination therapy and synthetic lethality should also be considered.

Genetic epistasis refers to gene-gene interactions that diversify cellular phenotypes, creating an assortment of cellular properties along the continuum of clonal expansion.55,163,164 Genomic instability alters the network of gene interactions during tumor progression, making epistasis an evolutionary process.98,165 Genetic epistasis and host immunosurveillance are responsible for inefficiency of metastatic colonization, where thousands of cells disseminate but only a rare few survive the metastatic cascade.16,166 Epistasis may also be an essential factor in the dormancy-to-proliferation switch, reactivating tumor dormancy.16 As mutations increase during cell transformation from normal to cancerous, gene interactions across the tumor-stromal axis evolve to maintain cell survival.150,163,165,166 Interactions that abrogate immunomodulation of the metastatic process in immunotherapy is not understood.16 Growing tumors adapt to microenvironmental changes during immunotherapy, and evolving interactions across the tumor-stromal axis prepare tumor cells for immunotherapeutic escape.167 This might happen through development of compound haploinsufficiencies, such as through SNPs and chromosomal microdeletions, which can be difficult to detect.135,168,169 For instance, regulatory T cells (Tregs) are immunosuppressive, and effector T cells (Teffs) are immunostimulatory. Tumor cells that recruit and respond to Teffs may succumb to anti-tumor immune responses, whereas tumor cells that recruit Tregs may prevail in their immunoescape.167 Treg recruitment may occur through unsuspecting mutation-induced compound haploinsufficiencies that suppress Teff recruitment and alter TSIs. Perhaps if such an “interactive switch” did not occur, immunoresponsiveness would prevail. Mutations endow reproductive fitness, and as a mutator phenotype, tumor cells might be able to force stabilized evolution through mutations to avert cell death. Much is still unknown, and nothing is conclusive. Recalling GOQ-1, the robust regularity that we see in LC and PC interpatient heterogeneity is the ability of cancerous cells to change epistatically without compromising survival.170 Thus, epistasis is a powerful force in cancer evolution.

Signaling pathway crosstalk is as much a form of epistasis as it is a form of functional redundancy. Tumor dormancy, and the seed and soil hypothesis, feature this integration of concepts. Dormancy is thought to occur by the seed and soil process in some cases, particularly in cases of immunosuppression and therapeutic resistance.16,28,75,171 The lynchpin is pathway crosstalk interactions across the tumor-stromal axis. Co-evolution of tumor and stromal cells occurs by interactions that constitutively evolve (epistasis) and constitutively signal (redundancy) along the progression continuum, inducing reactivation from dormancy.16,25,28,75,163,172 Intertumoral heterogeneity exhibits regularity by the evolving molecular interactions and changeful signaling pathways that occur across the tumor-stromal axis, case by case.123,173-176 Tumors harbor malignant cells with the evolvability of surviving the host immune response, metastatic cascade, and tumor dormancy.68 In every individual case, we observe cells attempting to survive, creating wounds that never heal. Tumor-stromal regulation must be unraveled to determine biomarkers that direct cancer cell evolution towards cell death, as depicted in spontaneous regression.167,177

Gene pleiotropy is also a factor in producing diverse cancer phenotypes. Gene pleiotropy refers to the influence of a gene (or pathway) on multiple phenotypic properties and mechanisms.178 Like redundancy and epistasis, gene pleiotropy significantly influences cancer behavior. From the stromal perspective, one of the biggest concerns about gene pleiotropy relates to chemokine and cytokine biology. Many chemokines and cytokines are characteristically pleiotropic.111,178‒182 Pleiotropic effects induce immunosuppression in the stroma, and increase phenotypic change in tumor cells at the invasion front.178,183 Mutations such as SNPs, microdeletions, microinsertions, and chromosomal rearrangements can induce pleiotropic effects in LC cells, which, in turn, destabilize phenotypic stability in stromal cells, and affects immunotherapeutic treatment.184 From the tumor perspective, pleiotropism triggers CSCs and other cancerous cells endowed with molecular fitness primed for evolvability toward the furtherance of disease - this is exhibited in the WNT signaling pathway.185 Aside from pleiotropy, tumor cell production of chemokines and cytokines also exhibit functional redundancy, which exacerbates the molecular complexity of tumor progression.181,182 Downregulation of a pleiotropic chemokine receptor, such as CXCR4, can produce phenotypic changes in an epithelial cancer cell without affecting its survival and progression potential. CXCR4 is implicated in numerous processes that cross between pleiotropy and redundancy, which serve to sustain, clone, and diversify cellular phenotypes. These molecular properties may justify why downregulation of CXCR4 impacts tumor dormancy.186,187 Several WNT target genes, such as IL-8, SOX9, CD44, AXIN2, MMP7, VEGF, and TDGF-1, exemplify, or are affected by genetic pleiotropy and redundancy. Some of these actions affect the cell cycle, cytoskeleton proteins, matrix metalloproteinases, and cell-matrix interactions.188‒193 Therefore, WNT pathway upregulation, downregulation, or abrogation through canonical and non-canonical channels significantly affects clonal diversity and cancer behavior.190 The Hh pathway (and NOTCH) may exhibit similar properties to the WNT pathway in LC and PC metastasis, through the targeting of VEGF, although the pathways present different gene targeting programs.194,195 For example, downstream Hh/Gli target genes such as FOXM1, TWIST2, ZEB1, ZEB2, VEGF, Ang2, and BMP4 have pleiotropic and redundancy characteristics like WNT, imposing on cell proliferation, decreased apoptosis, and metastasis; however, this is a different gene targeting program from the WNT pathway, but both pathways engage angiogenesis through VEGF.194,196,197 Abrogation or activation of pleiotropic pathways may mediate tumor cell evolution. Considering the number of genes in the human genome, diversity seems almost limitless. Thus, gene pleiotropy, like redundancy and epistasis, has a big influence on tumor cell evolvability and metastatic potential.150 There is also redundant pleiotropy. Exosome content dispersion, in conjunction with chemokine, cytokine, and ECM receptor signaling cascades, may produce redundant pleiotropic effects, as secreted exosomes from tumor cells may contain the same pleiotropic chemokines released from stromal cells.198,199

These effects - redundancy, epistasis, and pleiotropy - have thus far proven indefatigable in stopping LC and PC progression and metastasis. Recalling GOQ-2 and GOQ-3, why are LC and PC primaries metastasizing from one to the other respective organ on a frequential (regular) basis? Why do CUPs develop most often from lung and pancreatic primaries?21‒24

Tumor heterogeneity, emt/met processes, and regulatory tsis in cancer behavior: Genetic background directs origination, clonality, and metastatic progression of LC and PC, and signaling pathways are the conduits of that directorial script.128,147,150,200-202 Stochastic variations in SNPs, genomic rearrangements, epigenetic alterations, LOH, haploinsufficiency, and so forth, are thought to influence tumor heterogeneity and interpatient regularity.203 Hh, NOTCH, WNT, and CXCR4/CXCL12 transmute genomic and epigenomic programming into actionable tumor progression.121,161,204-207 Without an understanding of the divergent molecular TSIs that impose on cancer behavior, a one-size-fits-all therapeutic approach in treating LC and PC patients has a inherent probability of failure.208‒210 Analyses regarding genetic background should be considered in the context of tissue specificity, as some mutations may generate different phenotypes in different anatomic sites of human body.165,211,212 Genetic background in epithelial tumor cells contains only part of the explanation for tumor heterogeneity and cancer behavior. Genetic background in stromal cells also has a big influence, suggesting that TSIs are uniformly regulatory in cancer behavior.17,62,67,75,213 Regulatory biomarkers are localized throughout the neoplastic microenvironment and are induced by genetic background cues in tumor cells and stromal cells.67,81,84,150,151,213‒218 The invasive front serves as the junction for regulatory TSIs. Examination of genetic background and intercellular signaling may unveil regulatory biomarkers that regard GOQ-2 and GOQ-3. Tumor invasion is variably dynamic and thought to occur by EMT processes in some contexts.204,219‒223 However, the EMT is not the only program for tumor invasion.

The EMT is a conserved process that occurs primarily in human embryogenesis. It is a well-documented process in development and disease.17,42,206 ,222,224 The EMT is defined by a three-fold classification scheme: Type 1 (embryogenesis, organ development), Type 2 (tissue repair, wound healing), and Type 3 (loss of cell-cell adhesion and apical-basal cell polarity in tumor progression).222 During Type 3 EMT, cancerous cells dedifferentiate into CSCs, and transdifferentiate into mesenchymal-like stromal cells, jointly invading the stroma, and promoting dissemination and metastasis. MET is the reversal of these conversions at metastatic sites after dissemination.225,226 Changes in epithelial, mesenchymal and intermediary biomarkers, e.g., cell-cell junctions, serve to confirm the validity of EMT.204,224 However, amoeboid invasion, coordinated collective invasion, and cohort invasion are other programs for tumor cell invasion and tissue remodeling aside from EMT.204,223 The genetic basis of tumor invasion is questionable, and the EMT must be validated as to when it does and does not occur - that is to say, when it is and is not capable of occurring.53,206,227‒230 Essentially, incongruence between the EMT process and clinical interpretation of disease course justifies the study of plasticity and genetic background in lung and pancreatic preneoplasias. If drivers can induce epithelial cancerous cells to disseminate from preneoplasias, then the genetic background capable of programming stemness and dissemination from the preinvasive stage, as opposed to an EMT program, is highly intriguing.231,235

Reports show that the EMT process conduces tumor heterogeneity.159,206,236,237 Tumor budding and clonal diversion occurs at the invasive front through dedifferentiation and transdifferentiation of epithelial cells, which is partly where the controversy about EMT lies.152,238 Regulatory pathway interactions in clonal heterogeneity and plasticity is not completely understood. Distinguishing EMT-type tumor cells from mesenchymal stromal cells at the invasive front is enigmatic, and may require elucidation of pathway interactions to differentiate them, as they have similar morphological and marker characteristics.227,237 Hh, NOTCH and WNT pathways are the channels that communicate tumor invasion in the EMT process. Hh and NOTCH promote E-cadherin decreases, resulting in loss of cell adhesion, and motility.221 WNT plays a critical role between tumor and stroma by initiating tumor budding and driving PC cell stemness.152,238 During tumor budding processes, EMT-type cells become mesenchymal-like.152,238‒240 CSCs and EMT-type cells have similar properties, proliferate heterogenically at the invasive front, and form a stem cell niche with stromal components.14,110,137,152,165,241 EMT-type cells do not proliferate during invasion, but may be triggered to transdifferentiate for proliferation.152 The Hh pathway also plays a role in driving forward hypoxia-induced EMT processes.242 Hypoxia and metabolic stress (nutrient deprivation) induce the stem cell niche to initiate tumor invasion and drug resistance through tumor-stromal paracrine signaling.110,159,220,237,241,243‒245

Hypoxia refers to deficient oxygen (O2) levels that are lower than normal in tissue (pO2 < 30mmHg).246,247 Reduction of O2 levels in tumors occurs by heterogenic tumor proliferation.248,249 Hypoxia is a kind of signaling pathway that prompts HIF-1 to induce stromal mesenchymal cells to secrete lysyl oxidase (LOX), which signals tumor cells to undergo dedifferentiation to CSCs, and mediates bone marrow progenitor cell recruitment at premetastatic niches for metastasis.137,221,247,250,251 Detached tumor cells likely divert from this program when there is no premetastatic niche conducive for survival.138,252 Hypoxia also induces angiogenesis through HIF-1 activation of VEGF, and again, tumor cells likely divert from this hypoxia-driven program in cases of tumor and angiogenic dormancy.75,252 Metabolic stress imposes similar dysfunctions. Genetic instability and acidification of the stroma through the Warburg Effect severely stress the metabolic machinery of tumor cells, driving HIF-1 induced survival mechanisms.244,249 Interactive pathways become engaged in the evasion of apoptosis. Tumor cells interact with extracellular matrix (ECM) and stromal effectors through chemokine, cytokine, and ECM receptors.254,255 Stromal signaling cascades crosstalk with tumor signaling pathways and their effectors during EMT, including NOTCH, WNT, and CXCR4, as tumor cells attempt to evade apoptosis.255‒259

EMT reversal is thought to play a role in completing metastasis.260 Although less is known about the MET process, there is convincing evidence regarding its concept. After EMT, intravasation, dissemination, extravasation, and metastatic colonization in secondary organs, heterogenic growth begins (after a period of dormancy). After dormancy, the MET process begins, as morphohistological characteristics dedifferentiate backwards from CSCs to epithelial cancer cells.250,260 Recalling, GOQ-4, eventually MET lesions gain rudimentary vasculature through angiogenesis and a desmoplastic stroma, recapitulating their primaries anew - which could be important features in tracking CUPs.67,261 Critical aspects about the MET process are receptor biomarkers, changeful interactions that accompany those biomarkers, and de novo, unique mechanisms that occur in the MET process.262 Developmental signaling pathways also play a role in the MET-metastatic process. Upregulation of the NOTCH pathway is implicated in conversion (EMT), whereas downregulation of the NOTCH pathway is implicated in reversion (MET).91,93,263 The WNT pathway is a known “double inducer” that drives EMT at the primary invasive front, and MET in metastasis.260 CXCR4/CXCL12 knockdown inhibits angiogenesis.264 These examples exhibit the essentiality of recapitulated developmental signaling in MET-metastatic processes. Therefore, further investigating these pathways regarding the differentiable molecular genetics controlling EMT, and MET, should be considered.

The common thread linking tumor heterogeneity, EMT/MET, and metastasis is regulatory TSIs. It is long known, as Hanahan and Weinberg pointed out, that homeostatic “gatekeeper” regulation (apoptosis-driven) and cell-adhesion regulation (anti-invasion-driven) are jointly compromised as cancer hallmarks.65 Hypoxia inducement of EMT somehow diverts homeostatic TSI regulation, and the question remains, “where is the connection?” 265 Furthermore, in single epithelial tumor cell dissemination, the hypoxia inducement program is likely not employed in cases of dormancy, reactivation, proliferation, and metastasis.266‒269 Perhaps a pan-cancer expression profiling approach (reviewed in the next section) can provide some clues as to how hypoxia and metabolic stress variably drive dysregulation of cell adhesion and apoptosis, and, how a singly disseminated epithelial cancer cell induces dormancy, reactivation, and metastasis.270,271 As it stands, the molecular basis of TSI regulation is not completely understood.173 Respecting genetic background, tumor cell pathway interactions with stromal regulation may help explain organ-specific metastatic behavior between the lung and pancreas.128,272 Hh, NOTCH, WNT, CXCR4, their crosstalks, and interactions with the stroma, drive heterogenic clonality, immunosuppression, and metastasis.14,67,224,273 Regulatory TSIs induce interactive pathways through chemokines, growth factors, ECM proteins, proteases, and protease inhibitors, among other effectors.263,274,275

From the stromal perspective, Hh, NOTCH, and CXCR4 are involved in immunosuppression as co-regulated by immune cells.276‒278 As described by Tlsty and Coussens, “the stroma consists of cells and connective tissue that provide contextual framework for an organ or tissue…(and) contributes to cancer development through the release of soluble mediators that regulate cell proliferation, migration, angiogenesis, tissue remodeling, metabolism, and genomic integrity”.279 The essentiality is that stromal cells regulate tumor progression with tumor cells - their reciprocal interactions are thought to work as a unified regulatory unit, as they do in development and normal tissue homeostasis. Therefore, TSIs is where genetic background is the most relevant to cellular plasticity, tumor progression, and metastasis.128,206,250,274,279 An example of unified regulation is exemplified by the Hh pathway. The homeostatic stromal response to Hh-driven proliferation breaks tumor invasiveness, which implies that homeostatic stromal regulation is compromised in hypoxia-induced tumor invasion.242,276 Interestingly, experimental Hh pathway ablation during oncogenesis led to depletion of stromal fibroblasts, immunosuppression, induction of EMT, and tumor invasiveness, which explains why Hh inhibitors in clinical trials don’t work, and implies that the Hh pathway has some role in regulating the balance between epithelial and stromal elements.276,280,281 The WNT pathway drives forward tumor invasion and secondary metastasis at both invasive fronts; however, fibroblast-secreted exosomes intercommunicate with the WNT pathway to conduce cell polarity for this process to occur.198 NOTCH is involved in immunosuppression and metastatic-MET processes; however, NOTCH regulation of metastasis is induced by mesenchymal stromal cells that secrete the ligand Jagged2, which activates the NOTCH pathway, suppresses host immunity, and drives forward metastasis.277,281,282 The CXCR4/CXCL12 pathway is a driver of VEGF-mediated angiogenesis and recruits MDSCs and Tregs in immunosuppression;278,283 however, CXC4 and CXCL12 control of angiogenesis is regulated by cancer-associated fibroblasts that secrete IL-6 and VEGF.284 In these examples, we observe cooperative-stromal regulation. Thus, genetic background can abrogate or alter the regulatory effects from either side of the tumor-stromal interface.

From the tumor perspective, again, tumor regulation interacts with stromal regulation, and these processes operate in unison in tumor progression. Glycolytic breakdown during nutrient depravation produces lactic acid and carbonic acid, which are released from cancer cells, creating an acidic stroma. Low pH in the stroma co-opts the inflammatory response.285 Dysregulation of the inflammatory response is driven by TSIs and is a cancer hallmark.286 Tumor-released exosomes mediate stromal cell regulation of immunosuppression, angiogenesis, tumor aggressivity, and drug resistance.197,287,288 Only a small number of studies have looked at how developmental signaling pathways in tumor cells inter-regulate with the stroma tumor-secreted exosomes 289‒291Conceptually, such interactions should not be stones left unturned in research. More work on tumor-derived exosomes regarding recapitulated signaling and TSIs, could perhaps better explain the metastatic behaviors of lung and pancreatic lesions.

Pan-Cancer analysis: Pan-cancer analysis from the cancer genome atlas (TCGA) project is a bioinformatics and omics approach that employs databases used to extract information regarding effective biomarkers and other related cancer concerns.10,270 Used in conjunction with other technologies, assessments can unveil molecular clues about TSIs and the clonal evolution of cancerous cells. By searching for biomarkers across LC and PC subtypes, ranging in disease severity from preinvasive to aggressively metastatic, and then comparing those biomarkers through analyses with other cancers, clues could be discovered that can help explain cancer evolution and the connections between LC and PC metastatic behavior. Tumor-stromal-regulated immune responses, EMT and non-EMT tumor cell invasion, organ-specific metastases, rare metastasis, and intracellular biomarkers that mediate cancer cell evolution could also be explored. In such inquiries, stochastic similarities, dissimilarities, and patterns may be a common occurrence. The objective is to extract information regarding tumor-stromal regulation and cancer cell evolution that correlates with genomic displays across the tumor-stromal axis. Validation studies might confirm suspected biomarkers and their roles in cancer behavior, such as primary growth regression and angiogenic dormancy.

Genetic background across the tumor-stromal axis is the blueprint for cancer behavior.219,251,292 Inquiries might include: pleiotropic pathways, genomic reconfigurations (e.g., aneuploidy, copy number variations, microinsertions, microdeletions, and translocations), genetic mutations (e.g., promoter, intronic, exonic, and splicing mutations), epigenetic alterations (e.g., de-, hyper-, and hypo-methylation, and deacetylation), epistatic genes and their interactions, diversified miR expression, EMT and non-EMT drivers of tumor invasion, autophagy and metabolism, ECM interactions, exosome-content dispersion, inflammatory mediators, stromal and ECM mediators, and angiogenic and lymphangiogenic mediators. For example, the NOTCH pathway is sometimes ablated in lung SCCs through loss-of-function mutations.120 However, NOTCH mediates critical tumor-stromal communication.80 Queries regarding abrogation of NOTCH, and subsequent crosstalks and genomic changes in TSIs might lead to information regarding tumor cell evolution. Inquiries can also reveal fitness and selection by determining to what degree genetic redundancy is correlated with LC and PC behavior ranging from benign (e.g., cartilaginous hamartoma lung and serous cystic neoplasms pancreas) to malignant (e.g., SCLC and PDAC). Importantly, exome inquires may not entail all clues, as intronic and intergenic mutations, for example, also impact LC and PC biology.293,154

Inquiries could also be augmented with proteomics and molecular biology techniques to determine if there is specified regulation, such as for exosome-content dispersion.294 Suppression of immune cells (dendritic, NK-, and T-cells) through exosome communication plays an important role in tumor progression.217 In the lung, immunosuppression by alveolar macrophages is a normal physiological, homeostatic function, and it must be to handle the constant onslaught of antigens through breathing.295 Homeostatic balance between immunosuppression and immunoresponsiveness in the lung becomes dysregulated in tumor progression.148,163Cancer cell evolution and immune system evasion are reasons why immunotherapy is not always effective in treating LC patients, and the process is not biologically understood. In PC, TAMs are thought to be strong mediators of immunosuppression. Comparative biologic behavior across subtypes through pan-cancer, proteomics, and molecular biology analyses, among other techniques, may yield regulatory clues regarding exosome-content dispersion.

It is likely that pan-cancer analysis will be routinely used in medical treatment. Cancer behavior studies can accelerate the move forward. Memorial Sloan Kettering, for example, employs a next generation genomic sequencing test to determine if a patient’s cancer carries druggable mutations, and then matches that patient to specific therapies.296 However, even if LC or PC patients have druggable mutations, it is likely that drug resistance at some point will emerge. Other medical centers also operate genomics facilities. Cancer behavior studies may be the lynchpin that effectively integrates genomic analysis with efficacious medical treatment.

Discussion

In the pursuit to improve personalized medicine in metastatic LC and PC cases, perhaps we may not be seeing the forest for the trees. Essential molecular underpinnings may already be documented, including biomarkers in this review. Perhaps validation of research findings and formulation of remedies, although a slow process, is all that is needed to see remarkable change in LC and PC outcomes. However, if after such efforts there are still not enough biomarkers that can distinguish cancer behavior, then more work should be pinpointed in this area. Important biomarkers can emerge from organ-specific cancer behavior research. With respect to genetic background and uniform tumor-stromal regulation, biomarkers that can be exploited for efficacy is a main objective. Cancer behavior studies can capture the evolutionary underpinnings of what is presented for treatment to clinicians. Tumor-stromal regulation drives: (1) initiation toward primary tumor dormancy or indolency that remains undetectable for years; (2) dissemination of malignant cells from the primary site before localized growth, which may never become clinically detectable, causing CUPs or metastatic dormancy; and (3) localized tumor growth and progression that does not result in metastasis. For these reasons, studies that focus distinctly on cancer behavior across the full range of malignancy from preinvasion to metastasis should be carried out. Smoking reduction, for example, has resulted in declines in LC incidence; however, PC incidence has remained stagnant with a slight increase although smoking is a leading risk factor for both diseases, suggesting other inducers. Tobacco and alcohol use are high risk factors for head and neck cancer (HNC). Studies that examine benign and aggressive HNC cancer in patients who do and do not consume tobacco and alcohol may yield biomarkers. Other examples can be cited for melanoma and multiple myeloma. Dormant primary and micrometastatic tumors are commonly determined at autopsy, however, cancer behavior studies might enable clinicians to detect them in live patients. Furthermore, tumorigenic cells can disseminate during preneoplasia, but there are no validated biomarkers that indicate whether this likely has or has not occurred. Finally, primary site cancer cells can transform to dormant or indolent growth behavior before the primary site is detectable. Imaging and immunohistochemical methods determine unknown origins based on secondary malignancy, but the approach is problematic, and determinations are often arbitrarily made. Biomarkers that signify primary site (clonal origins) are needed, which is another reason why organ-specific metastatic research must continue.272,297,298

Conclusion

Advances made through an understanding of LC and PC metastatic behavior would include, but are not limited to: validation of biomarkers in cancer pre-initiation, initiation, and metastatic progression; validation of biomarkers that suggest primary site location and perioperative tumor dormancy; validation of molecular signatures of cancer risk in smokers; and implementation of population-based molecular and radiologic cancer screening. The first two of this series are subjects of this review. With respect to genetic background and tumor-stromal regulation, developmental signaling pathways that partly explain LC and PC metastatic behavior have been brought to light. The Hh pathway co-regulates preneoplastic signaling toward malignancy, CSC activation, EMT mechanisms, and induces drug resistance. The NOTCH pathway co-regulates EMT progression, tumor heterogeneity, CSC dormancy and reactivation, and, it induces metastasis, disease recurrence, and resistance to chemotherapy. The WNT pathway co-regulates CSC dormancy and reactivation, EMT processes, immunosuppression, and metastasis. The CXCL12/CXCR4 pathway co-regulates organ-specific homing of CSCs, organ-specific metastasis, immunosuppression, and the metastatic cascade. Hh, NOTCH, WNT and CXCL12/CXCR4 help drive metastasis across the tumor-stromal interface. Pan-cancer technology that can be used to help elucidate LC and PC metastatic behavior has also been brought to light. Databases can be referenced in research and treatment to aid personalized treatment approaches. In closing, research that investigates organ-specific metastasis (alongside rare metastatic occurrences) is warranted to enhance resolutions regarding cancer behavior. Perhaps such investigations could bring forth biomarkers indicative of early onset, diagnosis, and prognosis. Likewise, perhaps improved personalized medicine can be realized through differentiated treatment approaches based on cancer behavior studies.

Acknowledgments

None.

Conflicts of interest

Author declares there are no conflicts of interest.

References

  1. Cancer Facts and Figures 2017. American Cancer Society: Atlanta, USA; 2017.
  2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66(1):7‒30.
  3. 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(11):2913‒2921.
  4. Molina JR, Yang P, Cassivi SD, et al. Non-small cell lung cancer: epidemiology, risk factors, treatment, and survivorship. Mayo Clin Proc. 2008;83(5):584‒594.
  5. Sohn TA, Yeo CJ, Cameron JL, et al. Resected adenocarcinoma of the pancreas-616 patients: results, outcomes, and prognostic indicators. J Gastrointest Surg. 2000;4(6):567‒579.
  6. Deeb A, Haque SU, Olowokure O. Pulmonary metastases in pancreatic cancer, is there a survival influence? J Gastrointest Oncol. 2015;6(3):E48‒E51.
  7. Poruk KE, Wolfgang CL. Palliative management of unresectable pancreas cancer. Surg Oncol Clin N Am. 2016;25(2):327‒337.
  8. Ansari D, Gustafsson A, Andersson R. Update on the management of pancreatic cancer: surgery is not enough. World J Gastroenterol. 2015;21(11):3157‒3165.
  9. Richer AL, Friel JM, Carson VM, et al. Genomic profiling toward precision medicine in non-small cell lung cancer: getting beyond EGFR. Pharmgenomics Pers Med. 2015;8:63‒79.
  10. Cancer Genome Atlas Research Network, Weinstein JN, Collisson EA, et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet. 2013;45(10):1113‒1120.
  11. Klein CA. Parallel progression of primary tumours and metastases. Nat Rev Cancer. 2009;9(4):302‒312.
  12. Nguyen DX, Bos PD, Massagué J. Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer. 2009;9(4):274‒284.
  13. Ogino S, Fuchs CS, Giovannucci E. How many molecular subtypes? Implications of the unique tumor principle in personalized medicine. Expert Rev Mol Diagn. 2012;12(6):621‒628.
  14. Tsai JH, Yang J. Epithelial-mesenchymal plasticity in carcinoma metastasis. Genes Dev. 2013;27(20):2192‒2206.
  15. Hillerdal G. Indolent lung cancers--time for a paradigm shift: a review. J Thorac Oncol. 2008;3(3):208‒211.
  16. Sosa MS, Bragado P, Aguirre-Ghiso JA. Mechanisms of disseminated cancer cell dormancy: an awakening field. Nat Rev Cancer. 2014;14(9):611‒622.
  17. Mittal V. Epithelial mesenchymal transition in aggressive lung cancers. Adv Exp Med Biol. 2016;890:37‒56.
  18. Peeters CF, de Waal RM, Wobbes T, et al.Metastatic dormancy imposed by the primary tumor: does it exist in humans? Ann Surg Oncol. 2008;15(11):3308‒3315.
  19. Prasad V, Oseran A, Fakhrejahani F. The use of gene expression profiling and mutation analysis increases the cost of care for patients with carcinoma of unknown primary; does it also improve survival? Eur J Cancer. 2016;54:159‒162.
  20. Samadder NJ, Smith KR, Hanson H, et al. Familial risk in patients with carcinoma of unknown primary. JAMA Oncol. 2016;2(3):340‒346.
  21. Stella GM, Senetta R, Cassenti A, et al. Cancers of unknown primary origin: current perspectives and future therapeutic strategies. J Transl Med. 2012;10:12.
  22. Massard C, Loriot Y, Fizazi K. Carcinomas of an unknown primary origin--diagnosis and treatment. Nat Rev Clin Oncol. 2011;8(12):701‒710.
  23. Pentheroudakis G, Briasoulis E, Pavlidis N. Cancer of unknown primary site: missing primary or missing biology? Oncologist. 2007;12(4):418‒425.
  24. López-Lázaro M. The migration ability of stem cells can explain the existence of cancer of unknown primary site. Rethinking metastasis. Oncoscience. 2015;2(5):467‒475.
  25. Lqbal W, Alkarim S, AlHejin A, et al. Targeting signal transduction pathways of cancer stem cells for therapeutic opportunities of metastasis. Oncotarget. 2016;7(46):76337‒76353.
  26. Wood SL, Pernemalm M, Crosbie PA, et al. The role of the tumor-microenvironment in lung cancer-metastasis and its relationship to potential therapeutic targets. Cancer Treat Rev. 2014;40(4):558‒566.
  27.  , Lacobuzio Donahue CA. The pathology and genetics of metastatic pancreatic cancer. Arch Pathol Lab Med. 2009;133(3):413‒422.
  28. Talmadge JE, Fidler IJ. AACR centennial series: the biology of cancer metastasis: historical perspective. Cancer Res. 2010;70(14):5649‒5669.
  29. Hilbe W, Manegold C, Pircher A. Targeting angiogenesis in lung cancer - Pitfalls in drug development. Transl Lung Cancer Res. 2012;1(2):122‒128.
  30. Nicolson GL. Organ specificity of tumor metastasis: role of preferential adhesion, invasion and growth of malignant cells at specific secondary sites. Cancer Metastasis Rev. 1988;7(2):143‒188.
  31. Disibio G, French SW. Metastatic patterns of cancers: results from a large autopsy study. Arch Pathol Lab Med. 2008;132(6):931‒939.
  32. Sperti C, Moletta L, Patanè G. Metastatic tumors to the pancreas: The role of surgery, World J Gastrointest Oncol. 2014;6(10):381‒392.
  33. Gonlugur U, Mirici A, Karaayvaz M. Pancreatic involvement in small cell lung cancer. Radiol Oncol. 2014;48(1):11‒19.
  34. Adsay NV, Andea A, Basturk O, et al. Secondary tumors of the pancreas: an analysis of a surgical and autopsy database and review of the literature. Virchows Arch. 2004;444(6):527‒535.
  35. Fletcher, Christoper DM. Diagnostic Histopathology of Tumors. (4th edn), Churchill Livingstone Atlanta, Georgia, USA. 2013;pp.:296.
  36. Takaori K. Current understanding of precursors to pancreatic cancer. J Hepatobiliary Pancreat Surg. 2007;14(3):217‒223.
  37. Kauhanen S, Rinta Kiikka I, Kemppainen J, et al. Accuracy of 18F-FDG PET/CT, multidetector CT, and MR imaging in the diagnosis of pancreatic cysts: a prospective single-center study. J Nucl Med. 2015;56(8):1163‒1168.
  38. Krasinskas AM, Chiosea SI, Pal T, et al. KRAS mutational analysis and immunohistochemical studies can help distinguish pancreatic metastases from primary lung adenocarcinomas. Mod Pathol. 2014;27(2):262‒270.
  39. Alomari AK, Ustun B, Aslanian HR, et al. Endoscopic ultrasound-guided fine-needle aspiration diagnosis of secondary tumors involving the pancreas: An institution's experience. Cytojournal. 2016;13:1.
  40. Claire D, Marine G, Aurélie A, et al. Heterogeneity of metastatic pancreatic adenocarcinoma: Lung metastasis show better prognosis than liver metastasis-a case control study. Oncotarget. 2016;7(29):45649‒45655.
  41. Wangjam T, Zhang Z, Zhou XC, et al. Resected pancreatic ductal adenocarcinomas with recurrence limited in lung have a significantly better prognosis than those with other recurrence patterns. Oncotarget. 2015;6(34):36903‒36910.
  42. Yamashita K, Miyamoto A, Hama N, et al. Survival impact of pulmonary metastasis as recurrence of pancreatic ductal adenocarcinoma. Dig Surg. 2015;32(6):464‒471.
  43. Berx G, Raspé E, Christofori G, et al. Pre-EMTing metastasis? Recapitulation of morphogenetic processes in cancer. Clin Exp Metastasis. 2007;24(8):587‒597.
  44. Jennings RE, Berry AA, Strutt JP, et al. Human pancreas development. Development. 2015;142(18):3126‒3137.
  45. Swarr DT, Morrisey EE. Lung endoderm morphogenesis: gasping for form and function. Annu Rev Cell Dev Biol. 2015;31:553‒573.
  46. Cheng X, Ying L, Lu L, et al. Self-renewing endodermal progenitor lines generated from human pluripotent stem cells. Cell Stem Cell. 2012;10(4):371‒384.
  47. Cagle, Philip T, Allen Timothy C, et al. Advances in Surgical Pathology: Lung Cancer. Arch Pathol Lab Med. 2011;135(1):110‒116.
  48. Ishizumi T, McWilliams A, MacAulay C, et al. Natural history of bronchial preinvasive lesions. Cancer Metastasis Rev. 2010;29(1):5‒14.
  49. Cooper CL, O'Toole SA, Kench JG. Classification, morphology and molecular pathology of premalignant lesions of the pancreas. Pathology. 2013;45(3):286‒304.
  50. Lam S, Szabo E. Preinvasive endobronchial lesions: lung cancer precursors and risk markers? Am J Respir Crit Care Med. 2013;192(12):1411‒1413.
  51. Benson RE, Rosado de Christenson ML, Martínez Jiménez S, et al. Spectrum of pulmonary neuroendocrine proliferations and neoplasms. Radiographics. 2013;33(6):1631‒1649.
  52. Jang KT, Park SM, Basturk O, et al. Clinicopathologic characteristics of 29 invasive carcinomas arising in 178 pancreatic mucinous cystic neoplasms with ovarian-type stroma: implications for management and prognosis. Am J Surg Pathol. 2015;39(2):179‒187.
  53. Biankin AV, Kench JG, Dijkman FP, et al. Molecular pathogenesis of precursor lesions of pancreatic ductal adenocarcinoma. Pathology. 2003;35(1):14‒24.
  54. Christiansen JJ, Rajasekaran AK. Reassessing epithelial to mesenchymal transition as a prerequisite for carcinoma invasion and metastasis. Cancer Res. 2006;66(17):8319‒8326.
  55. Merrick DT, Gao D, Miller YE, et al. Persistence of bronchial dysplasia is associated with development of invasive squamous cell carcinoma. Cancer Prev Res (Phila). 2016;9(1):96‒104.
  56. Yachida S, Lacobuzio Donahue CA. Evolution and dynamics of pancreatic cancer progression. Oncogene. 2013;32(45):5253‒5260.
  57. Pipinikas CP, Kiropoulos TS, Teixeira VH, et al. Cell migration leads to spatially distinct but clonally related airway cancer precursors. Thorax. 2014;69(6):548‒557.
  58. Yachida S, Jones S, Bozic I, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467(7319):1114‒1117.
  59. Hsiong SX, Carampin P, Kong HJ, et al. Differentiation stage alters matrix control of stem cells. J Biomed Mater Res A. 2008;85(1):145‒156.
  60. Liu X, Pitarresi JR, Cuitiño MC, et al. Genetic ablation of Smoothened in pancreatic fibroblasts increases acinar-ductal metaplasia. Genes Dev. 2016;30(17):1943‒1955.
  61. Freire J, Ajona D, De Biurrun G, et al. Silica-induced chronic inflammation promotes lung carcinogenesis in the context of an immunosuppressive microenvironment. Neoplasia. 2013;15(8):913‒924.
  62. Hutchinson L. Genetics: CUP: discovering genetic opportunities. Nat Rev Clin Oncol. 2015;12(5):251.
  63. Saadi A, Shannon NB, Lao-Sirieix P, et al. Stromal genes discriminate preinvasive from invasive disease, predict outcome, and highlight inflammatory pathways in digestive cancers. Proc Natl Acad Sci USA. 2010;107(5):2177‒2182.
  64. Kurahashi I, Fujita Y, Arao T, et al. A microarray-based gene expression analysis to identify diagnostic biomarkers for unknown primary cancer. PLoS One. 2013;8(5):e63249.
  65. Pentheroudakis G, Spector Y, Krikelis D, et al. Global microRNA profiling in favorable prognosis subgroups of cancer of unknown primary (CUP) demonstrates no significant expression differences with metastases of matched known primary tumors. Clin Exp Metastasis. 2013;30(4):431‒439.
  66. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100(1):57‒70.
  67. Atlasi Y, Looijenga L, Fodde R. Cancer stem cells, pluripotency, and cellular heterogeneity: a WNTer perspective. Curr Top Dev Biol. 2014;107:373‒404.
  68. Aiello NM, Bajor DL, Norgard RJ, et al. Metastatic progression is associated with dynamic changes in the local microenvironment. Nat Commun. 2016;7:12819.
  69. Horak E, Darling DL, Tarin D. Analysis of organ-specific effects on metastatic tumor formation by studies in vitro. J Natl Cancer Inst . 1986;76(5):913‒922.
  70. Książkiewicz M, Markiewicz A, Zaczek AJ. Epithelial-mesenchymal transition: a hallmark in metastasis formation linking circulating tumor cells and cancer stem cells. Pathobiology. 2012;79(4):195‒208.
  71. Newman K, Stahl-Herz J, Kabiawu O, et al. Pancreatic carcinoma with multilineage (acinar, neuroendocrine, and ductal) differentiation. Int J Clin Exp Pathol. 2009;2(6):602‒607.
  72. Lantuéjoul S, Salameire D, Salon C, et al. Pulmonary preneoplasia-sequential molecular carcinogenetic events. Histopathology. 2009;54(1):43‒54.
  73. Gazdar AF, Brambilla E. Preneoplasia of lung cancer. Cancer Biomark. 2010;9(1-6):385‒396.
  74. Killen H. DIPNECH presenting on a background of malignant melanoma: new lung nodules are not always what they seem. BMJ Case Reports bcr. 2014;2014:203667.
  75. Pelosi G, Fabbri A, Cossa M, et al. What clinicians are asking pathologists when dealing with lung neuroendocrine neoplasms? Semin Diagn Pathol. 2015;32(6):469‒479.
  76. Műzes G, Sipos F. Metastatic cell dormancy and re-activation: an overview on series of molecular events critical for cancer relapse. Anticancer Agents Med Chem. 2016.  
  77. Wilkerson MD, Yin X, Walter V, et al. Differential pathogenesis of lung adenocarcinoma subtypes involving sequence mutations, copy number, chromosomal instability, and methylation. PLoS One. 2012;7(5):e36530.
  78. Sakashita S, Sakashita M, Sound Tsao M. Genes and pathology of non-small cell lung carcinoma. Seminars in Oncology. 2014;41(1):28‒39.
  79. Moffitt RA, Marayati R, Flate EL, et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet. 2015;47(10):1168‒1178.
  80. Borczuk AC, Gorenstein L, Walter KL, et al. Non-small-cell lung cancer molecular signatures recapitulate lung developmental pathways. Am J Pathol. 2003;163(5):1949‒1960.
  81. Takebe N, Miele L, Harris PJ, et al. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat Rev Clin Oncol. 2015;12(8):445‒464.
  82. Ylipää A, Yli-Harja O, Zhang W, Characterization of aberrant pathways across human cancers. BMC Syst Biol. 2013;7 Suppl1: S1.
  83. Lomberk G, Urrutia R . Primers on molecular pathways--Notch. Pancreatology. 2008;8(2):103‒104.
  84. Garnis C, Campbell J, Davies JJ, et al. Involvement of multiple developmental genes on chromosome 1p in lung tumorigenesis. Hum Mol Genet. 2005;14(4):475‒482.
  85. Stewart DJ. Wnt signaling pathway in non-small cell lung cancer. J Natl Cancer Inst. 2014;106(1):djt356.
  86. Vaz AP, Ponnusamy MP, Seshacharyulu P, et al. A concise review on the current kunderstanding of pancreatic cancer stem cells. J Cancer Stem Cell Res. 2014;2. pii:e1004.
  87. Akinleye A, Iragavarapu C, Furqan M, et al. Novel agents for advanced pancreatic cancer. Oncotarget. 2015;6(37):39521‒39537.
  88. Watkins DN, Berman DM, Burkholder SG, et al. Hedgehog signalling within airway epithelial progenitors and in small-cell lung cancer. Nature. 2003; 422(6929):313‒317.
  89. Lin EH, Kao YR, Lin CA, et al. Hedgehog pathway maintains cell survival under stress conditions, and drives drug resistance in lung adenocarcinoma. Oncotarget. 2016;7(17): 24179‒24193.
  90. Hao K, Tian XD, Qin CF, et al. Hedgehog signaling pathway regulates human pancreatic cancer cell proliferation and metastasis. Oncol Rep. 2013;29(3):1124‒1132.
  91. Takebe N, Miele L, Harris PJ, et al. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat Rev Clin Oncol. 2015;12(8):445‒464.
  92. Ni X, Long J, Cen P, et al. Pancreatic cancer tumour initiating cells: the molecular regulation and therapeutic values. J Cell Mol Med. 2012;16(5): 988‒994.
  93. Xie M,Zhang L, He CS, et al. Activation of Notch-1 enhances epithelial-mesenchymal transition in gefitinib-acquired resistant lung cancer cells. J Cell Biochem. 2012;113(5):1501‒1513.
  94. Hassan KA, Wang L, Korkaya H, et al. Notch pathway activity identifies cells with cancer stem cell-like properties and correlates with worse survival in lung adenocarcinoma. Clin Cancer Res. 2013;19(8):1972‒1980.
  95. Sahin IH, Iacobuzio-Donahue CA, O'Reilly EM. Molecular signature of pancreatic adenocarcinoma: an insight from genotype to phenotype and challenges for targeted therapy. Expert Opin Ther Targets. 2016;20(3):341‒59.
  96. Collu GM, Hidalgo Sastre A, Brennan K. Wnt-Notch signalling crosstalk in development and disease. Cell Mol Life Sci. 2014;71(18):3553‒3567.
  97. Takebe N, Miele L, Harris PJ, et al. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat Rev Clin Oncol. 2015;12(8):445‒464.
  98. Reya T, Clevers H. Wnt signalling in stem cells and cancer. Nature. 2005;434(7035):843‒850.
  99. Giancotti FG. Mechanisms governing metastatic dormancy and reactivation. Cell. 2013;155(4):750‒764.
  100. Pollard JW. Defining metastatic cell latency. N Engl J Med. 2016;375(3):280‒282.
  101. Malladi S, Macalinao DG, Jin X, et al. Metastatic latency and immune evasion through autocrine inhibition of WNT. Cell. 2016;165(1):45‒60.
  102. Wang Z, Ma Q, Liu Q, et al. Blockade of SDF-1/CXCR4 signalling inhibits pancreatic cancer progression in vitro via inactivation of canonical Wnt pathway. Br J Cancer. 2008;99(10):1695‒1703.
  103. Li X, Ma Q, Xu Q, et al. SDF-1/CXCR4 signaling induces pancreatic cancer cell invasion and epithelial-mesenchymal transition in vitro through non-canonical activation of Hedgehog pathway. Cancer Lett. 2012;322(2):169‒176.
  104. Cui J, Jiang W, Wang S, et al. Role of Wnt/β-catenin signaling in drug resistance of pancreatic cancer. Curr Pharm Des. 2012;18(17):2464‒2471.
  105. Kleffel S, Schatton T. Tumor dormancy and cancer stem cells: two sides of the same coin? Adv Exp Med Biol. 2013;734:145‒179.
  106. Bleau AM, Agliano A, Larzabal L, et al. Metastatic dormancy: a complex network between cancer stem cells and their microenvironment. Histol Histopathol. 2014;29(12):1499‒1510.
  107. Wang Z, Dabrosin C, Yin X, et al. Broad targeting of angiogenesis for cancer prevention and therapy. Semin Cancer Biol. 2015;35(Suppl):S224‒S243.
  108. Wald O, Shapira OM, Izhar U. CXCR4/CXCL12 axis in non-small cell lung cancer (NSCLC) pathologic roles and therapeutic potential. Theranostics. 2013;3(1):26‒33.
  109. Wu PF, Lu ZP, Cai BB, et al. Role of CXCL12/CXCR4 signaling axis in pancreatic cancer. Chin Med J (Engl). 2013;126(17):3371‒334.
  110. Villasenor A, Cleaver O. Crosstalk between the developing pancreas and its blood vessels: an evolving dialog. Semin Cell Dev Biol. 2012;23(6):685‒692.
  111. Borovski T, De Sousa E Melo F, Vermeulen L, et al. Cancer stem cell niche: the place to be. Cancer Res. 2011;71(3):634‒639.
  112. Ben Baruch A. Organ selectivity in metastasis: regulation by chemokines and their receptors. Clin Exp Metastasis. 2008;25(4):345‒356.
  113. Heiler S, Wang Z, Zöller M. Pancreatic cancer stem cell markers and exosomes - the incentive push. World J Gastroenterol. 2016;22(26):5971‒6007.
  114. Gassmann P, Haier J, Schlüter K, et al. CXCR4 regulates the early extravasation of metastatic tumor cells in vivo. Neoplasia. 2009; 11(7):651‒661.
  115. Kitamura T, Qian BZ, Pollard JW. Immune cell promotion of metastasis. Nat Rev Immunol. 2015;15(2):73‒86.
  116. Zou W. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer. 2005;5(4):263‒274.
  117. Pawig L, Klasen C, Weber C, et al. Diversity and inter-connections in the CXCR4 chemokine receptor/ligand family: molecular perspectives. Front Immunol. 2015;6:429.
  118. Subbannayya T, Variar P, Advani J, et al. An integrated signal transduction network of macrophage migration inhibitory factor. J Cell Commun Signal. 2016;10(2):165‒170.
  119. Matthaios D, Zarogoulidis P, Balgouranidou I, et al. Molecular pathogenesis of pancreatic cancer and clinical perspectives. Oncology. 2011;81(3-4):259‒272.
  120. Kure S, Matsuda Y, Hagio M, et al. Expression of cancer stem cell markers in pancreatic intraepithelial neoplasias and pancreatic ductal adenocarcinomas. Int J Oncol. 2012;41(4):1314‒1324.
  121. Wang NJ, Sanborn Z, Arnett KL, et al. Loss-of-function mutations in Notch receptors in cutaneous and lung squamous cell carcinoma. Proc Natl Acad Sci USA. 2011;108(43):17761‒17766.
  122. Karamboulas C, Ailles L. Developmental signaling pathways in cancer stem cells of solid tumors. Biochim Biophys Acta. 2013;1830(2):2481‒2495.
  123. Nussinov R, Tsai CJ, Mattos C. 'Pathway drug cocktail': targeting Ras signaling based on structural pathways. Trends Mol Med. 2013;19(11):695‒704.
  124. Vogelstein B, Papadopoulos N, Velculescu VE, et al. Cancer genome landscapes. Science. 2013;339(6127):1546‒1558.
  125. Deininger P. Genetic instability in cancer: caretaker and gatekeeper genes. Ochsner J. 1999;1(4):206‒209.
  126. Loeb LA. Human cancers express mutator phenotypes: origin, consequences and targeting. Nature Rev Cancer. 2011;11(6):450‒457.
  127. Troche JR, Mayne ST, Freedman ND, et al. The Association Between Alcohol Consumption and Lung Carcinoma by Histological Subtype. Am J Epidemiol. 2016;183(2):110‒121.
  128. Duell EJ. Epidemiology and Potential Mechanisms of Tobacco Smoking and Heavy Alcohol Consumption in Pancreatic Cancer. Mol Carcinog. 2012;51(1):40‒52.
  129. Ponder BA. Cancer genetics. Nature. 2001;411(6835):336‒341.
  130. Singhi AD, Ishida H, Ali SZ, et al. A Histomorphologic Comparison of Familial and Sporadic Pancreatic Cancers. Pancreatology. 2015;15(4):387‒391.
  131. Tersmette AC, Petersen GM, Offerhaus GJ, et al. Increased Risk of Incident Pancreatic Cancer Among First-degree Relatives of Patients with Familial Pancreatic Cancer. Clin Cancer Res. 2001;7(3):738‒744.
  132. Moslein G, Tester DJ, Lindor NM, et al. Microsatellite instability and mutation analysis of hMSH2 and hMLH1 in patients with sporadic, familial and hereditary colorectal cancer. Hum Mol Genet. 1996; 5(9):1245‒1252.
  133. Devilee P, Cleton Jansen AM, Cornelisse CJ. Ever since Knudson. Trends Genet. 2001;17(10):569‒573.
  134. Morton JP, Jamieson NB, Karim SA, et al. LKB1 haploinsufficiency cooperates with Kras to promote pancreatic cancer through suppression of p21-dependent growth arrest. Gastroenterology. 2010;139(2):586‒597.
  135. Celeste A, Difilippantonio S, Difilippantonio MJ, et al. H2AX haploinsufficiency modifies genomic stability and tumor susceptibility. Cell. 2003;114:371‒383.
  136. Balmain A, Gray J, Ponder B. The genetics and genomics of cancer. Nat Genet. 2003;33 Suppl:238‒244.
  137. Fidler IJ. The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nature Rev Cancer. 2003;3(6):453‒458.
  138. Chang AL, Schwertschkow AH, Nolta JA, et al. Involvement of mesenchymal stem cells in cancer progression and metastases. Curr Cancer Drug Targets. 2015;15(2):88‒98.
  139. Sceneay J, Smyth MJ, Möller A. The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 2013;32(3-4):449‒464.
  140. Jeon JS, Zervantonakis IK, Chung S, et al. In vitro model of tumor cell extravasation. PLoS One. 2013;8(2):e56910.
  141. Nicolson GL. Cancer metastasis: tumor cell and host organ properties important in metastasis to specific secondary sites. Biochim Biophys Acta. 1988;948(2):175‒224.
  142. Hu JB, Jin M, Chen EG, Lung squamous cell carcinoma metastasizing to the nasopharynx following bronchoscopy intervention therapies: a case report. World J Surg Oncol. 2014;12: 68.
  143. Damonte P, Hodgson JG, Chen JQ, et al. Mammary carcinoma behavior is programmed in the precancer stem cell. Breast Cancer Res. 2008;10(3): R50.
  144. Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science. 2011;331(6024):1559‒1564.
  145. Cheng G. A case of lung squamous cell carcinoma with duodenal metastasis on FDG PET/CT. Clin Nucl Med. 2016;41(8): 659‒660.
  146. Delitala AP, Vidili G, Manca A, et al. A case of thyroid metastasis from pancreatic cancer: case report and literature review. BMC Endocr Disord. 2014;14:6.
  147. Martinez P, Birkbak NJ, Gerlinger M, ,et al. Parallel evolution of tumour subclones mimics diversity between tumours. J Pathol . 2013;230(4):356‒364.
  148. Burrell RA, McGranahan N, Bartek J, et al. The causes and consequences of genetic heterogeneity in cancer evolution. Nature. 2013;501(7467):338‒345.
  149. Pan S, Dong Q, Sun LS, et al. Mechanisms of Inactivation of PTCH1 Gene in Nevoid Basal Cell Carcinoma Syndrome: Modification of the Two-Hit Hypothesis. Clin Cancer Res. 2010;16(2):442‒450.
  150. Pavelic SK, Sedic M, Bosnjak H, et al. Metastasis: new perspectives on an old problem. Mol Cancer. 2011;10:22.
  151. Greaves M, Maley CC. Clonal evolution in cancer. Nature. 2012;481(7381):306‒313.
  152. Visvader J. Cells of origin in cancer. Nature. 2011;469(7330):314‒322.
  153. Karamitopoulou E. Tumor budding cells, cancer stem cells and epithelial-mesenchymal transition-type cells in pancreatic cancer. Front Oncol. 2013;2:209.
  154. Salk JJ, Fox EJ, Loeb LA. Mutational Heterogeneity in Human Cancers: Origin and Consequences. Annu Rev Pathol. 2010;5:51‒75.
  155. Tahira AC, Kubrusly MS, Faria MF, et al. Long noncoding intronic RNAs are differentially expressed in primary and metastatic pancreatic cancer. Mol Cancer. 2011;10:141.
  156. Diss G, Ascencio D, DeLuna A, et al. Molecular mechanisms of paralogous compensation and the robustness of cellular networks. J Exp Zool B Mol Dev Evol. 2014;322(7):488‒499.
  157. Espinosa-Cantú A, Ascencio D, Barona-Gómez F, et al. Gene duplication and the evolution of moonlighting proteins. Front Genet. 2015;6:227.
  158. Cereda M, Mourikis TP, Francesca D, et al. Genetic redundancy, functional compensation, and cancer vulnerability. Trends in Cancer. 2016;2(4):160‒162.
  159. Trang SH, Joyner DE, Damron TA, et al. Potential for functional redundancy in EGF and TGFalpha signaling in desmoid cells: a cDNA microarray analysis. Growth Factors. 2010;28(1):10‒23.
  160. Zhang J, Tian XJ, Xing J. Signal transduction pathways of EMT induced by TGF-β, SHH, and WNT and their crosstalks. J Clin Med. 2016;5:4.
  161. Rueff J, Rodrigues AS. Cancer drug resistance: a brief overview from a genetic viewpoint. Methods Mol Biol. 2016;1395:1‒18.
  162. Javelaud D, Pierrat MJ, Mauviel A. Crosstalk between TGF-β and hedgehog signaling in cancer. FEBS Lett. 2012;586(14):2016‒2025.
  163. Holohan C, Van Schaeybroeck S, Longley DB, et al. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer. 2013;13(10):714‒726.
  164. Casbas Hernandez P, Fleming JM, Troester MA. Gene expression analysis of in vitro cocultures to study interactions between breast epithelium and stroma. J Biomed Biotechnol. 2011;2011:520987.
  165. Sackton TB, Hartl DL. Genotypic context and epistasis in individuals and populations. Cell. 2016;166(2):279‒287.
  166. Ashworth A, Lord CJ, Reis-Filho JS. Genetic interactions in cancer progression and treatment. Cell. 2011;145(1):30‒38.
  167. Malanchi I, Santamaria Martınez A, Susanto E, et al. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature. 2012;481(7379):85‒89.
  168. Hegmans JP, Aerts JG. Immunomodulation in cancer. Current Opinion in Pharmacology. 2014;17:17‒21.
  169. Manikandan M, Raksha G, Munirajan AK. Haploinsufficiency of tumor suppressor genes is driven by the cumulative effect of microRNAs, microRNA binding site polymorphisms and microRNA polymorphisms: An in silico approach. Cancer Inform. 2012;11:157‒71.
  170. Huang N, Lee I, Marcotte EM, et al. Charact erising and Predict ing Haploins ufficiency in the Human Genome. PLoS Genet. 2010;6(10):e1001154.
  171. Makohon Moore A, Iacobuzio Donahue CA. Pancreatic cancer biology and genetics from an evolutionary perspective. Nat Rev Cancer. 2016;16(9):553‒565.
  172. Massagué J, Obenauf AC. Metastatic colonization by circulating tumour cells. Nature. 2016;529(7586):298‒306.
  173. Vogelstein B, Kinzle KW. Cancer genes and the pathways they control. Nat Med. 2004;10(8):789‒799.
  174. LaFoya B, Munroe JA, Mia MM, et al. Notch: A multi-functional integrating system of microenvironmental signals. Dev Biol. 2016;418(2):227‒241.
  175. Mimeault M, Batra SK. Altered gene products involved in the malignant reprogramming of cancer stem/progenitor cells and multitargeted therapies. Mol Aspects Med. 2014;39:3‒32.
  176. Jones S, Zhang X, Parsons DW, et al. Core Signaling Pathways in Human Pancreatic Cancers Revealed by Global Genomic Analyses. Science. 2008;321(5897):1801‒1806.
  177. Kreeger PK, Lauffenburger DA. Cancer systems biology: a network modeling perspective. Carcinogenesis. 2009;31(1):2‒8.
  178. Saito T, Naito M, Matsumura Y, et al. Spontaneous regression of a large hepatocellular carcinoma with multiple lung metastases. Gut Liver. 2014;8(5):569‒574.
  179. Neuzillet C, Tijeras-Raballand A, Cohen R, et al. Targeting the TGF-β pathway for cancer therapy. Pharmacol Ther. 2015;147:22‒31.
  180. Ratajczak MZ, Zuba-Surma E, Kucia M, et al. The pleiotropic effects of the SDF-1-CXCR4 axis in organogenesis, regeneration and tumorigenesis. Leukemia. 2006;20(11):1915-1924.
  181. Feurino LW, Fisher WE, Bharadwaj U, et al. Current Update of Cytokines in Pancreatic Cancer: Pathogenic Mechanisms, Clinical Indication, and Therapeutic Values. Cancer Invest. 2006;24(7):696‒703.
  182. Ozaki K, Leonard WJ. Cytokine and cytokine receptor pleiotropy and redundancy. J Biol Chem. 2002;277(33):29355‒29358.
  183. Amedei A, Prisco D, D Elios MM. The use of cytokines and chemokines in the cancer immunotherapy. Recent Pat Anticancer Drug Discov. 2013;8(2):126‒142.
  184. Burkholder B, Huang RY, Burgess R, et al. Tumor-induced perturbations of cytokines and immune cell networks. Biochim Biophys Acta. 2014;1845(2):182‒201.
  185. Rizvi NA, Hellmann MD, Snyder A, et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348(6230):124‒128.
  186. Yang K, Wang X, Zhang H, et al. The evolving roles of canonical WNT signaling in stem cells and tumorigenesis: implications in targeted cancer therapies. Lab Invest . 2015;96(2):116‒136.
  187. Cojoc M, Peitzsch C, Trautmann F, et al. emerging targets in cancer management: role of the CXCL12/CXCR4 axis. Onco targets Ther. 2013;6:1347‒1361.
  188. Nobutani K, Shimono Y, Mizutani K, et al. Downregulation of CXCR4 in metastasized breast cancer cells and implication in their dormancy. PLoS One. 2015;10(6):e0130032.
  189. Macheda ML, Stacker SA. Importance of Wnt signaling in the tumor stroma microenvironment. Curr Cancer Drug Targets. 2008;8(6):454‒465.
  190. Larsimont JC, Youssef KK, Sánchez-Danés A, et al. Sox9 Controls Self-Renewal of Oncogene Targeted Cells and Links Tumor Initiation and Invasion. Cell Stem Cell. 2015;17(1):60‒73.
  191. Zhan T, Rindtorff N, Boutros M. Wnt signaling in cancer. Oncogene; 2016.
  192. Overall CM, Dean RA. Degradomics: Systems biology of the protease web. Pleiotropic roles of MMPs in cancer. Cancer Metastasis Rev. 2006;25(1):69‒75.
  193. Sedgwick AE, D Souza Schorey C. Wnt signaling in cell motility and invasion: drawing parallels between development and cancer. Cancers (Basel). 2016;8(9):80.
  194. Sellerio AL, Ciusani E, Ben-Moshe NB, et al. Overshoot during phenotypic switching of cancer cell populations. Sci Rep. 2015;5:15464.
  195. Zhang X, Gaspard JP, Chung DC. Regulation of vascular endothelial growth factor by the Wnt and K-ras pathways in colonic neoplasia. Cancer Res. 2001;61(16):6050‒6054.
  196. Wang Z, Li Y, Kong D, et al. Cross-talk between miRNA and Notch signaling pathways in tumor development and progression. Cancer Lett . 2010;292(2):141‒148.
  197. Weinberg RA., Twisted epithelial-mesenchymal transition blocks senescence. Nat Cell Biol. 2008;10(9):1021‒1023.
  198. Ruzinova MB, Benezra R. Id proteins in development, cell cycle and cancer. Trends Cell Biol. 2003;13(8):410‒418.
  199. Iero M, Valenti R, Huber V, et al. Tumour-released exosomes and their implications in cancer immunity. Cell Death Differ. 2008;15(1):80‒88.
  200. Luga V, Wrana JL. Tumor-stroma interaction: revealing fibroblast-secreted exosomes as potent regulators of Wnt-planar cell polarity signaling in cancer metastasis. Cancer Res. 2013;73(23):6843‒6847.
  201. Semenova EA, Nagel R, Berns A. Origins, genetic landscape, and emerging therapies of small cell lung cancer. Genes Dev. 2015;29(14):1447‒1462.
  202. Sandhu V, Wedge DC, Bowitz Lothe IM, et al. The Genomic Landscape of Pancreatic and Periampullary Adenocarcinoma. Cancer Res. 2016;76(17):5092‒5102.
  203. Gilbertson RJ. Mapping cancer origins. Cell. 2011;145(1):25‒29.
  204. Rübben A, Nordhoff O. A systems approach defining constraints of the genome architecture on lineage selection and evolvability during somatic cancer evolution. Biol Open. 2013;2(1):49‒62.
  205. Friedl P, Alexander S. Cancer invasion and the microenvironment: plasticity and reciprocity. Cell. 2011;147(5):992‒1009.
  206. Zoccoli A, Iuliani M, Pantano F, et al. Premetastatic niche: ready for new therapeutic interventions? Expert Opin Ther Targets 16 Suppl. 2012;2:S119‒S129.
  207. Yang J, Weinberg RA. Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell. 2008;14(6):818‒829.
  208. Fortini ME. Notch signaling: the core pathway and its posttranslational regulation. Dev Cell. 2009;16:633‒647.
  209. Gottesman MM, Lavi O, Hall MD, et al. Toward a better understanding of the complexity of cancer drug resistance. Annu Rev Pharmacol Toxicol. 2016;56:85‒102.
  210. Hay M, Thomas DW, Craighead JL, et al. Clinical development success rates for investigational drugs. Nat Biotechnol. 2014;32(1):40‒51.
  211. Kamb A, Wee S, Lengauer C. Why is cancer drug discovery so difficult? Nature Rev Drug Discov. 2007;6(2):115‒120.
  212. Sakurai H, Asamura H, Miyaoka E, et al. Differences in the prognosis of resected lung adenocarcinoma according to the histological subtype: a retrospective analysis of Japanese lung cancer registry data. Eur J Cardiothorac Surg. 2014;45(1):100‒107.
  213. Cohen RL, Settleman J. From cancer genomics to precision oncology-tissue’s still an issue. Cell. 2014;157:1509‒1514.
  214. Bidard FC, Pierga JY, Vincent-Salomon A, et al. A "class action" against the microenvironment: do cancer cells cooperate in metastasis? Cancer Metastasis Rev. 2008;27(1):5‒10.
  215. Coulson Thomas YM, Gesteira TF, Norton AL, et al. The role of proteoglycans in the reactive stroma on tumor growth and progression. Histol Histopathol. 2015;30(1):33‒41.
  216. Dotto GP. Multifocal epithelial tumors and field cancerization: stroma as a primary determinant. J Clin Invest. 2014;124(4):1446‒1453.
  217. McGranahan N, Swanton C. Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell. 2015;27(1):15‒26.
  218. Whiteside TL. Exosomes and tumor-mediated immune suppression. J Clin Invest. 2016;126(4):1216‒1223.
  219. Payne KK, Keim RC, Graham L, et al. Tumor-reactive immune cells protect against metastatic tumor and induce immunoediting of indolent but not quiescent tumor cells. J Leukoc Biol. 2016;100(3):625‒635.
  220. Yamaguchi H, Sakai R. Direct interaction between carcinoma cells and cancer associated fibroblasts for the regulation of cancer invasion. Cancers (Basel). 2015;7(4):2054‒2062.
  221. Wu Y, Zhou BP. Inflammation: a driving force speeds cancer metastasis. Cell Cycle. 2009;8(20):3267‒3273.
  222. Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 2014;15(3):178‒196.
  223. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;119(6):1420‒1428.
  224. Yilmaz M, Christofori G, Lehembre F. Distinct mechanisms of tumor invasion and metastasis. Trends Mol Med. 2007;13(12):535‒541.
  225. Nieto MA, Huang RY, Jackson RA, et al.EMT: 2016. Cell . 2016;116(1):21‒45.
  226. Singh A, Settleman J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene. 2010;29(34):4741‒4751.
  227. Huang Z, Wu T, Liu AY, et al. Differentiation and transdifferentiation potentials of cancer stem cells. Oncotarget. 2015;6(37):39550‒39563.
  228. Chui MH. Insights into cancer metastasis from a clinicopathologic perspective: epithelial-mesenchymal transition is not a necessary step. Int J Cancer. 2013;132(7):1487‒1495.
  229. Zheng X, Carstens JL, Kim J, et al. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature. 2015;527(7579):525‒530.
  230. Tarin D, Thompson EW, Newgreen DF. The fallacy of epithelial mesenchymal transition in neoplasia. Cancer Res. 2005;65(14):5996‒6000.
  231. Zhao M, Kong L, Liu Y, et al. dbEMT: an epithelial-mesenchymal transition associated gene resource. Sci Rep. 2015;5:11459.
  232. Thompson LD, Becker RC, Adair CF, et al. Mucinous cystic neoplasm (mucinous cystadenocarcinoma of low-grade malignant potential) of the pancreas. Am J Surg Pathol . 1999;23(1):1‒16.
  233. Kargozaran H, Vu V, Ray P, et al. Invasive IPMN and MCN: same organ, different outcomes? Ann Surg Oncol. 2011;18(2):345‒351.
  234. Kerr KM. Pulmonary preinvasive neoplasia. J Clin Pathol. 2001;54(4):257‒271.
  235. Ooi AT, Gower AC, Zhang KX, et al. Molecular profiling of premalignant lesions in lung squamous cell carcinomas identifies mechanisms involved in stepwise carcinogenesis. Cancer Prev Res (Phila). 2014;7(5):487‒495.
  236. Selamat SA, Galler JS, Joshi AD, et al. DNA methylation changes in atypical adenomatous hyperplasia, adenocarcinoma in situ, and lung adenocarcinoma. PLoS One. 2011;6(6):e21443.
  237. Sato R, Semba T, Saya H, et al. Concise review: stem cells and epithelial-mesenchymal transition in cancer: biological implications and therapeutic targets. Stem Cells. 2016;34(8):1997‒2007.
  238. Gao D, Vahdat LT, Wong S, et al. Microenvironmental regulation of epithelial-mesenchymal transitions in cancer. Cancer Res. 2012;72(19):4883‒4889.
  239. De Craene B, Berx G. Regulatory networks defining EMT during cancer initiation and progression. Nature Rev Cancer. 2013;13:97‒110.
  240. Kalluri R. EMT: when epithelial cells decide to become mesenchymal-like cells. J Clin Invest. 2009;119(6):1417‒1419.
  241. Polyak K, Weinberg RA. Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nat Rev Cancer. 2009;9:265‒273.
  242. Irmisch A, Huelsken J. Metastasis: new insights into organ-specific extravasation and metastatic niches. Exp Cell Res. 2013;319(11):1604‒1610.
  243. Lei J, Ma J, Ma Q, et al. Hedgehog signaling regulates hypoxia induced epithelial to mesenchymal transition and invasion in pancreatic cancer cells via a ligand-independent manner. Mol Cancer. 2013;12:66.
  244. Shi Q, Abbruzzese JL, Huang S, et al. Constitutive and inducible interleukin 8 expression by hypoxia and acidosis renders human pancreatic cancer cells more tumorigenic and metastatic. Clin Cancer Res. 1999;5(11):3711‒3721.
  245. Lu H, Forbes RA, Verma A. Hypoxia-inducible factor 1 activation by aerobic glycolysis implicates the Warburg Effect in carcinogenesis. J Biol Chem. 2002;277(26):23111‒23115.
  246. Gort EH, Groot AJ, van der Wall E, et al. Hypoxic regulation of metastasis via hypoxia-inducible factors. Curr Mol Med. 2008;8(1):60‒67.
  247. Jiang J, Tang Y, Liang X. EMT: A new vision of hypoxia promoting cancer progression. Cancer Biol Ther. 2011;11(8): 714‒723.
  248. Hill RP, Marie Egyptienne DT, Hedley DW. Cancer stem cells, hypoxia, and metastasis. Semin Radiat Oncol . 2009;19:106‒111.
  249. Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nat Rev Cancer. 2011;11(6):393‒410.
  250. Harris AL. Hypoxia - a key regulatory factor in tumour growth. Nat Rev Cancer. 2002;2(1):38‒47.
  251. Varga J, De Oliveira T, Greten FR. The architect who never sleeps: tumor-induced plasticity. FEBS Lett. 2014;588(15):2422‒2427.
  252. Erler JT, Weaver VM. Three-dimensional context regulation of metastasis. Clin Exp Metastasis. 2009;26(1):35‒49.
  253. Duda DG, Jain RK. Premetastatic lung "niche": is vascular endothelial growth factor receptor 1 activation required? Cancer Res. 2010;70(14):5670‒5673.
  254. Weis SM, Cheresh DA. Tumor angiogenesis: molecular pathways and therapeutic targets. Nat Med. 2011;17(11):1359‒1370.
  255. Marcucci F, Bellone M, Caserta CA, et al. Pushing tumor cells toward a malignant phenotype: stimuli from the microenvironment, intercellular communications, and alternative roads. Int J Cancer. 2014;135(6):1265‒1276.
  256. Paoli P, Giannoni E, Chiarugi P. Anoikis molecular pathways and its role in cancer progression. Biochim Biophys Acta. 2013;1833(12):3481‒3498.
  257. Lindsey S, Langhans SA. Crosstalk of oncogenic signaling pathways during epithelial-mesenchymal transition. Front Oncol. 2014;4:358.
  258. Taparra K, Tran PT. Hijacking the hexosamine biosynthetic pathway to promote EMT-mediated neoplastic phenotypes. Front Oncol. 2016;6:85.
  259. Wang X, Fu AQ, McNerney ME, et al. Widespread genetic epistasis among cancer genes. Nat Commun. 2014;5:4828.
  260. Moustakas A, Heldin P. TGFβ and matrix-regulated epithelial to mesenchymal transition. Biochim Biophys Acta. 2014;1840(8):2621‒2634.
  261. Yao D, Dai C, Peng S. Mechanism of the mesenchymal-epithelial transition and its relationship with metastatic tumor formation. Mol Cancer Res. 2011;9(12):1608‒1620.
  262. Soltermann A, Tischler V, Arbogast S, et al. Prognostic significance of epithelial-mesenchymal and mesenchymal-epithelial transition protein expression in non-small cell lung cancer. Clin Cancer Res. 2008;14(22):7430‒7437.
  263. Gao D, Joshi N, Choi H, et al. Myeloid progenitor cells in the premetastatic lung promote metastases by inducing mesenchymal to epithelial transition. Cancer Res. 2012;72(6):1384‒1394.
  264. Wang Z, Li Y, Kong D, Banerjee S, et al. Acquisition of epithelial-mesenchymal transition phenotype of gemcitabine-resistant pancreatic cancer cells is linked with activation of the notch signaling pathway. Cancer Res. 2009;69(6):2400‒2407.
  265. Katkoori VR, Basson MD, Bond VC, et al. Nef-M1, a peptide antagonist of CXCR4, inhibits tumor angiogenesis and epithelial-to-mesenchymal transition in colon and breast cancers. Oncotarget. 2015;69(29):27763‒27777.
  266. Bao B, Azmi AS, Ali S, et al. The biological kinship of hypoxia with CSC and EMT and their relationship with deregulated expression of miRNAs and tumor aggressiveness. Biochim Biophys Acta. 2012;1826(2):272‒296.
  267. Coghlin C, Murray G. The role of gene regulatory networks in promoting cancer progression and metastasis. Future Onco. 2014;l 10(5):735‒748.
  268. Klein CA. Selection and adaptation during metastatic cancer progression. Nature. 2013;501(7467):365‒372.
  269. Billaud M, Santoro M. Is Co-option a prevailing mechanism during cancer progression? Cancer Res. 2011;71(21):6572‒6575.
  270. Sleeman JP, Christofori G, Fodde R, et al. Concepts of metastasis in flux: the stromal progression model. Semin Cancer Biol. 2012;22(3):174‒186.
  271. Knijnenburg TA, Bismeijer T, Wessels LF, et al. A multilevel pan-cancer map links gene mutations to cancer hallmarks. Chin J Cancer. 2015;34(10):439‒449.
  272. Nath A, Chan C. Genetic alterations in fatty acid transport and metabolism genes are associated with metastatic progression and poor prognosis of human cancers. Sci Rep. 2016;6:18669.
  273. Bellovin DI, Das B, Felsher DW. Tumor dormancy, oncogene addiction, cellular senescence, and self-renewal programs. Adv Exp Med Biol. 2013;734:91‒107.
  274. Ilmer M, Horst D. Pancreatic CSCs and microenvironment. Genes Cancer. 2015;6(9-10):365‒366.
  275. Joyce JA, Pollard JW. Microenvironmental regulation of metastasis. Nat Rev Cancer. 2009;9(4):239‒252.
  276. Hu M, Polyak K. Microenvironmental regulation of cancer development. Curr Opin Genet Dev. 2008;18(1):27‒34.
  277. Lee JJ, Perera RM, Wang H, et al. Stromal response to Hedgehog signaling restrains pancreatic cancer progression. Proc Natl Acad Sci U S A. 2014;111(30):E3091‒E3100.
  278. Del Papa B, Sportoletti P, Cecchini D, et al. Notch1 modulates mesenchymal stem cells mediated regulatory T-cell induction. Eur J Immunol. 2013;43(1):182‒187.
  279. Obermajer N, Muthuswamy R, Odunsi K, et al. PGE2-induced CXCL12 production and CXCR4 expression controls the accumulation of human MDSCs in ovarian cancer environment. Cancer Res. 2011;71(24):7463‒7470.
  280. Tlsty TD, Coussens LM. Tumor stroma and regulation of cancer development. Annu Rev Pathol. 2006;1:119‒150.
  281. Neesse A, Algül H, Tuveson DA, et al. Stromal biology and therapy in pancreatic cancer: a changing paradigm. Gut. 2015;64(9):1476‒1484.
  282. Özdemir BC, Pentcheva Hoang T, Carstens JL, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25(6):719‒734.
  283. Yang Y, Ahn YH, Gibbons DL, et al. The Notch ligand Jagged2 promotes lung adenocarcinoma metastasis through a miR-200-dependent pathway in mice. J Clin Invest. 2011;121(4):1373‒1385.
  284. Liang Z, Brooks J, Willard M, et al. CXCR4/CXCL12 axis promotes VEGF-mediated tumor angiogenesis through Akt signaling pathway. Biochem Biophys Res Commun. 2007;359(3):716‒722.
  285. Nagasaki T, Hara M, Nakanishi H, et al. Interleukin-6 released by colon cancer-associated fibroblasts is critical for tumour angiogenesis: anti-interleukin-6 receptor antibody suppressed angiogenesis and inhibited tumour-stroma interaction. Br J Cancer. 2014;110(2):469‒478.
  286. Wang T, Liu G, Wang R. The Intercellular Metabolic Interplay between Tumor and Immune Cells. Front Immunol . 2014;5:358.
  287. Colotta F, Allavena P, Sica A, et al.Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis. 2009;30(7):1073‒1081.
  288. Becker A, Thakur BK, Weiss JM, et al. Extracellular Vesicles in Cancer: Cell-to-Cell Mediators of Metastasis. Cancer Cell. 2016;30(6): 836‒848.
  289. Kalluri R. The biology and function of exosomes in cancer. J Clin Invest. 2016;126(4):1208‒1215.
  290. Beloribi Djefaflia S, Siret C, Lombardo D. Exosomal lipids induce human pancreatic tumoral MiaPaCa-2 cells resistance through the CXCR4-SDF-1α signaling axis. Oncoscience. 2014;2(1):15‒30.
  291. Dovrat S, Caspi M, Zilberberg A, et al. 14-3-3 and β-catenin are secreted on extracellular vesicles to activate the oncogenic Wnt pathway. Mol Oncol. 2014;8(5):894‒911.
  292. Ristorcelli E, Beraud E, Mathieu S, et al. Essential role of Notch signaling in apoptosis of human pancreatic tumoral cells mediated by exosomal nanoparticles. Int J Cancer. 2009;125(5):1016‒1026.
  293. Lomberk GA, Urrutia R. The Triple-Code Model for Pancreatic Cancer: Cross Talk Among Genetics, Epigenetics, and Nuclear Structure. Surg Clin North Am. 2015;95(5):935‒952.
  294. Shultz JC, Goehe RW, Murudkar CS, et al. SRSF1 regulates the alternative splicing of caspase 9 via a novel intronic splicing enhancer affecting the chemotherapeutic sensitivity of non-small cell lung cancer cells. Mol Cancer Res. 2011;9(7):889‒900.
  295. Ricklefs F, Mineo M, Rooj AK, et al. Extracellular vesicles from high-grade glioma exchange diverse pro-oncogenic signals that maintain intratumoral heterogeneity. Cancer Res. 2016;76(10):2876‒2881.
  296. Schneider DJ, Speth JM, Peters-Golden M. Signed, Sealed, Delivered: Microenvironmental Modulation of Extracellular Vesicle-Dependent Immunoregulation in the Lung. Front Cell Dev Biol. 2016;4:94.
  297. Cheng DT, Mitchell TN, Zehir A, et al. Memorial Sloan Kettering-integrated mutation profiling of actionable cancer targets (MSK-IMPACT): A hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J Mol Diagn. 2015;17(3):251‒264.
  298. Coussens LM, Zitvogel L, Palucka AK. Neutralizing Tumor-Promoting Chronic Inflammation: A Magic Bullet? Science. 2013;339(6117):286‒291.
  299. Kristiansen S, Nielsen D, Sölétormos G. Methylated DNA for monitoring tumor growth and regression: how do we get there? Crit Rev Clin Lab Sci. 2014;51(3):149‒159.
Creative Commons Attribution License

©2017 Wilson. This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.

Citations