Next Article in Journal
Right Ventricle and Epigenetics: A Systematic Review
Previous Article in Journal
The Role of Autophagy in Type 2 Diabetic Kidney Disease Management
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

PAK in Pancreatic Cancer-Associated Vasculature: Implications for Therapeutic Response

by
Arian Ansardamavandi
1,
Mehrdad Nikfarjam
1,2 and
Hong He
1,*
1
Department of Surgery, Austin Precinct, The University of Melbourne, 145 Studley Rd, Heidelberg, VIC 3084, Australia
2
Department of Hepatopancreatic-Biliary Surgery, Austin Health, 145 Studley Rd, Heidelberg, VIC 3084, Australia
*
Author to whom correspondence should be addressed.
Cells 2023, 12(23), 2692; https://doi.org/10.3390/cells12232692
Submission received: 25 October 2023 / Revised: 20 November 2023 / Accepted: 22 November 2023 / Published: 23 November 2023
(This article belongs to the Section Cell Microenvironment)

Abstract

:
Angiogenesis has been associated with numbers of solid tumours. Anti-angiogenesis drugs starve tumours of nutrients and oxygen but also make it difficult for a chemo reagent to distribute into a tumour, leading to aggressive tumour growth. Anti-angiogenesis drugs do not appear to improve the overall survival rate of pancreatic cancer. Vessel normalisation is merging as one of the new approaches for halting tumour progression by facilitating the tumour infiltration of immune cells and the delivery of chemo reagents. Targeting p21-activated kinases (PAKs) in cancer has been shown to inhibit cancer cell growth and improve the efficacy of chemotherapy. Inhibition of PAK enhances anti-tumour immunity and stimulates the efficacy of immune checkpoint blockades. Inhibition of PAK also improves Car-T immunotherapy by reprogramming the vascular microenvironment. This review summarizes current research on PAK’s role in tumour vasculature and therapeutical response, with a focus on pancreatic cancer.

1. Introduction

The global incidence of pancreatic ductal adenocarcinoma (PDA) is increasing. Pancreatic cancer now ranks among the top ten most prevalent cancers [1]. The poor prognosis of PDA is mainly due to the lack of an early detection method and no effective systemic therapy for metastatic and locally advanced diseases [2]. Less than a third of patients receive surgical resection, of which 65% of patients experience tumour recurrence after surgical resection [3].
One of the main barriers to developing effective treatments for PDA is its unique characteristic of the tumour microenvironment (TME). The TME of PDA exhibits a dense stromal environment, containing cancer-associated fibroblasts and immune cells that interact with different subsets of cancerous cells, including cancer stem cells (CSCs) [4]. In addition, the blood vessels in the TME facilitate the exchange of nutrients, oxygen, and other essential factors and provide pathways for the dissemination of tumour cells to distant sites [5]. Interactions between blood vessels and other elements of the TME influence overall tumour behaviour and therapeutic response [6].
Inhibiting blood vessels may limit the expansion of the tumour and enhance the survival of cancer patients [7,8]. In 2004, FDA approved the clinical application of bevacizumab, an anti-VEGF antibody, to be used as an initial therapeutic approach for metastatic colorectal cancer (5-FU) [9]. Furthermore, FDA has approved multi-target tyrosine kinase inhibitors (TKIs) like sunitinib, sorafenib, and pazopanib. These TKIs are designed to specifically target VEGF receptors, with a particular focus on VEGFR-2 receptor [10]. In pancreatic cancer treatment, various clinical trials have been conducted to assess the efficacy of anti-angiogenic agents. However, the outcomes have been disappointing across multiple studies [11]. Although a few clinical trials have shown improvements in progression-free survival (PFS) [12], none of them have indicated any significant extension in the overall survival (OS) of pancreatic cancer patients. The use of anti-angiogenic treatment for pancreatic cancer remains a subject of debate [13]. Nevertheless, an increasing body of clinical evidence suggests that a substantial number of tumours exhibit an initial lack of response to anti-angiogenic agents, termed de novo resistance, while others gradually acquire resistance over time, resulting in tumour progression even after several months of treatment [14]. Although high microvessel density (MVD) is generally considered to be a predictor of poor prognosis [15,16], several studies found no correlation between poor prognosis and high MVD [17,18]. PDA exhibits limited response to traditional chemotherapy treatments because of the low MVD. Instead of the inhibition of blood vessels, vascular normalisation holds potential promise as an innovative approach for cancer treatment [19], especially for pancreatic cancer.
Over 90% of PDA carry KRas mutations [20]. KRas is a member of the Ras family proteins that bind to guanosine triphosphate (GTP), becoming active. When mutated, KRas becomes constitutively active, triggering the activation of downstream pathways critical for cell survival and growth [21]. Through direct and indirect mechanisms, KRas activates p21-activated kinases (PAKs) [22]. PAKs have gained considerable attention for their roles in the tumorigenesis of PDA [23]. Recent studies have also revealed PAK’s involvement in tumour vascular networks and immune response. In this review, we will explore the potential connections between PAK, vascularisation, and its impact on therapeutical response in pancreatic cancer.

2. Vascular Network and Therapeutic Responsiveness

Tumour blood vessels exhibit many defects, such as abnormal vessel growth, leaky structure with an absence of proper pericyte support, and loss of the normal structures found in arterioles, capillaries, and venules [24]. These defects of tumour blood vessels compromise perfusion, which in turn leads to a hypoxic and acidic microenvironment within the tumour [25]. These factors and/or events interact with each other (Figure 1) [9] and collectively hinder the effective delivery of chemotherapeutic agents [26], contributing to tumour growth, survival, and metastasis.
In the TME of pancreatic cancer, the accumulation of fibrotic stroma attributes to a robust desmoplastic response which increases the interstitial fluid pressure [28]. The elevated interstitial pressure compresses stromal blood vessels leading to hypovascular and hypoxic microenvironments [29], which may contribute to the unsatisfactory outcomes observed in anti-VEGF treatment [17]. In murine models of pancreatic neuroendocrine carcinoma and glioblastoma, angiogenesis inhibitors targeting the VEGF pathway have been shown to partially inhibit tumour growth, while causing increased invasiveness and elevated metastasis [30]. The increased invasiveness and metastasis triggered by anti-angiogenic treatment have also been observed in other types of tumours [31,32,33,34]. Furthermore, certain angiogenesis inhibitors, particularly broad-spectrum TKIs, have the capacity to concomitantly affect MVD and microvessel integrity (MVI), which decrease pericyte coverage around the blood vessels [34]. The reduction in coverage of vascular pericytes leads to the destabilisation of vessels, resulting in increased permeability and promoting metastasis [35]. Conversely, the vascular normalisation approach induces a more physiologically balanced state within the TME, resulting in better outcomes of patients with reduced adverse effects [36]. More importantly, the vascular normalisation approach contributes to optimising the effectiveness of radiotherapy, chemotherapy, and immunotherapy, thereby holding potential to bring about more favourable clinical outcomes [37] (Figure 2).
Preclinical studies have shown that tumour vasculature normalisation decreased hypoxia and thus metastasis while increasing drug delivery to the tumour site, enhancing the anti-tumour effect of the drug [38]. Tumour vasculature normalisation has been shown to enhance the anti-tumour immune response in other cancers [39,40]. Semaphorin 3A (SEMA3A) contributes to the physiological vascular normalisation via the negative regulation of integrins [41]. In pancreatic cancer, inhibiting STAT3 along with gemcitabine effectively increased MVD and facilitated the delivery of gemcitabine to tumours without changes in the stromal collagen or hyaluronan levels [42]. In breast cancer, Salvianolic acid B (SalB) can normalise blood vessels by stimulating the interactions between tumour cells and endothelial cells. As a result, cisplatin was delivered more effectively into the tumour, and tumour metastasis was decreased [43].
Histidine-rich glycoprotein (HRG), a highly prevalent and extensively characterised protein in the plasma [44], facilitated the stimulation of anti-tumour immune responses and the normalisation of blood vessels, which in turn decreased tumour growth and metastasis and increased the efficacy of chemotherapy [45]. In a separate study, a regulator of G-Protein Signalling 5 (RGS5), a major gene responsible for aberrant vascular architecture in cancer [46,47], triggered the maturation of pericytes, leading to vasculature normalisation and subsequently, reductions in both tumour hypoxia and vessel permeability. These modifications in the vasculature of the TME enhanced the influx of immune effector cells into the tumour, resulting in a substantial extension of mouse survival [47]. Table 1 provides a summary of the research studies conducted on diverse molecular targets aimed at normalising blood vessels in cancer.
The TME of pancreatic cancer contains an inefficient and leaky vasculature associated with poor tumour perfusion, leading to hypoxia and tumour metastasis. The failure of anti-angiogenesis drugs in the treatment of pancreatic cancer has urged the approach of tumour vasculature normalisation. PAKs have played important roles in endothelial cell proliferation, migration, and permeability, as well as in angiogenesis in cancer settings [48,49]. In the subsequent section, we will present an overview of the roles of PAKs in PDA-associated vasculature and the implication in therapeutic responses.
Table 1. Molecules used to normalise vessels and their effects and outcomes.
Table 1. Molecules used to normalise vessels and their effects and outcomes.
CancerTreatmentEffectResultRef.
PancreaticInhibition of STAT3+ gemcitabine↑ The number of blood vessels (CD31).
↓ Cytidine deaminase (cda) expression.
↓ Collagen alignment.
≈ Hyaluronan and collagen density.
↓ Tumour weight. ↑ Mouse survival.[42]
BreastSalvianolic acid B+ cisplatin↑ αSMA/CD31 area, NG2/CD31 area (pericyte coverage), Col IV/CD31 area.
↑ VE-cadherin/CD31 area, Lectin, and O2 average.
↓ Dextran%, PIMO, and HIF-1α.
↓ Hypoxia.
↓ Metastasis.
↑ Chemotherapy and drug penetration to the tumour.
[43]
Hepatocellular HRG via downregulation of placental growth factor (PIGF)↑ Vessel perfusion (Lectin + CD31 + Vessels).
↓ Haemorrhage.
↑ Pericyte coated vessels.
↓ Hypoxia.
↓ Tumour cell apoptosis.
TAM polarisation
↑ T cells (CD8+), dendritic cells (CD11c+), and NK cells.
↓ Necrosis.
[45]
Hepatocellular Tanshinone IIA↑ Tube formation.
≈ Microvessel density (MVD).
↑ Microvessel integrity (NG2 expression).
↓ Metastasis.[50]
BreastSinomenine hydrochloride↑ Lectin+CD31+vessels and αSMA positive vessels (pericyte coverage).
↓ HIF-1α.
↓ Hypoxia.
↓ Necrotic area.
↓ Growth rate and metastasis.
[51]
PancreaticInhibition of Rgs5↑ Number of vessels.
↓ Vessel diameter.
↑ αSMA and NG2 (pericyte coverage).
↓ Hypoxia.
↑ CD8 T cells.
↑ Mouse survival.
[47]

3. PAK in Pancreatic Cancer

PDA is dominated by mutations in the oncogene of KRas, which activates downstream signalling pathways including other GTPases such as CDC42 and Rac [52], which in turn activate PAKs [53]. There are two groups containing six members of PAKs: group 1—PAK1, PAK2, and PAK3; group 2—PAK4, PAK5, and PAK6. Alterations in the expression or activity of these PAK family members have been implicated in various physiological and pathological processes, including cell proliferation, migration and invasion, autophagy, immune cells activation, chemotherapy sensitivity, and survival [54].
The most extensively studied members among the PAK proteins are PAK1 and PAK4, which have been found to be overexpressed in many types of cancers [55,56,57]. The involvement of PAK1 in PDA tumorigenesis is characterised by its intricate and multifaceted nature. PAK1 interacts with the NFκB-p65 complex, thereby enhancing the transcriptional expression of fibronectin, causing cells to transform, and promoting the invasive epithelial–mesenchymal transition (EMT) [58]. PAK1 influences the production of VEGF, a growth factor that supports tumour invasion and metastasis [59,60].
PAK4 stimulates cell proliferation and survival through AKT and ERK pathways and plays a key role in regulating the proliferation of vascular smooth muscle cells (VSMCs) through the AKT pathway [61]. PAK4 activates the WNT signalling pathway via direct interaction with β-catenin, contributing to an immunosuppressive TME [62]. PAK4 also interacts with p85α, a PI3K regulatory component involved in cell migration. Inhibiting PAK4 or disrupting its connection with p85α reduces migration in pancreatic cancer cells [63,64]. PAK4 influences genes linked to stem cell traits, affecting tumour sphere formation through STAT3. Inhibiting PAK4 or STAT3 reduces stem cell-like characteristics in cancer cells, offering potential therapeutic options [65]. The details of PAKs’ function in pancreatic cancer have been reviewed elsewhere. We have provided a summary of PAK roles in pancreatic cancer broadly in Figure 3.
The unique dense stromal which exists in the TME of PDA compresses vasculatures and reduces blood vessel function, preventing the effective delivery of drugs into tumour sites and inhibiting the immune cell penetration to the tumour site. The regulation of tumour vasculature will have a significant impact on the efficacy of chemo- and immune-therapies. PAKs have recently gained attention in angiogenesis and the modulation of vasculature in cancer settings, including PDA [66,67,68,69]. Below, we will discuss the role of PAK 1 and PAK4 in tumour vasculature.

4. PAK and Tumour Vasculature

Rho, Rac, and Cdc42, members of the Rho family of small GTPases, are responsible for the rearrangement of actin cytoskeleton in cells and regulate integrin adhesion complexes located at the cell surface in response to various signals [70]. They also play integral roles in governing endothelial cell dynamics, (including proliferation, polarisation, intercellular adhesion, and migration), vascular permeability, and angiogenesis [71,72]. It has been shown that mice with endothelial-specific CDC42 knockout could not survive until birth owing to the lack of formation of blood vessels during the development of embryos, indicating the important role of CDC42 in the development of microvessels and vascular permeability [73]. Endothelial cells depleted of CDC42 formed a smaller tube, and the number of tubes formed and branching points were also decreased [74]. PAKs acting downstream of CDC42 have become the focus of research on the regulation and/or modulation of vasculature, particularly in cancer settings.
PAK1 has a vital role in the process of tumour angiogenesis. PAK1 amplification has been associated with advanced tumour stages, higher MVD, and notable colony stimulating factor 2 expression (CSF2) in myxofibrosarcoma. CSF2 is a cytokine that regulates the differentiation of macrophages and promotes angiogenesis, which is downregulated in PAK1 knockdown groups [75]. In breast cancer, PAK1 has been recognised as an essential element to stimulate VEGF production following the activation of human epidermal growth factor receptor signalling by Heregulin β1 (HRG). This process promotes the formation of new blood vessels [76]. The activation of PAK1 in a human cholangiocarcinoma cell impedes the degradation of hypoxia-inducible factor 1 alpha (HIF-1α) protein. This leads to the accumulation of HIF-1α, which in turn triggers the elevation of VEGF, consequently stimulating the process of tumour angiogenesis [77,78]. In a xenografted model of prostate cancer, inhibition of PAK1 decreased tumour growth, which was associated with a 70% reduction in laminin-positive blood vessels. This reduction in vascular density was associated with a tenfold decrease in matrix metalloproteinases 9 (MMP9) expression [79]. VEGF and angiogenesis are directly associated with the expression of MMPs [80].
In the TME of pancreatic cancer, there is a positive correlation between PAK1 expression and the expression of α-SMA and Desmin, suggesting the potential involvement of cancer-associated fibroblasts (CAFs), which are known contributors to angiogenesis [81]. PAK1 protein expression levels have been found to correlate positively with αSMA and Desmin in pancreatic stellate cells (PSCs) [82], which stimulate angiogenesis and facilitate tumour extravasation. These PSCs accompany circulating cancer cells, creating a vital microenvironment that facilitates primary cancer cells in establishing metastatic colonies [83]. Our preliminary data from a comprehensive global proteomic analysis showed a substantial decrease in fibronectin levels in the PAK1 knockout (KO) pancreatic cancer cells. This finding is of significant interest as fibronectin plays a pivotal role in promoting angiogenesis, a process crucial for the advancement of PDA [84].
PAK4 plays a critical role in coordinating the abnormal development of blood vessels and the exacerbation of low-oxygen (hypoxic) conditions within a tumour. In a kinome-wide screening of mesenchymal-like transcriptional activation in human glioblastoma-derived endothelial cells, PAK4 was identified as a selective regulator of genetic reprogramming in tumour vasculature, leading to aberrant vascularisation [85]. PAK4 knockout recovered the expression of adhesion proteins in tumour-derived endothelial cells, leading to normalisation of vasculature, which was associated with increased T cell infiltration and decreased tumour growth. PAK4 was found to reprogram the endothelial transcriptome and downregulate claudin-14 and VCAM-1 expression, enhancing vessel permeability and reducing T cell adhesion to the endothelium [85]. PAK4-regulated endothelial cell plasticity contributes to the modification of the vascular microenvironment and thus the efficacy of cancer therapy.
In prostate cancer, inhibition of PAK4 increased the width of blood vessels in the tumour context [86]. Furthermore, the blood vessel density stained by PECAM1 (CD31) showed a trend of elevation in the PAK4 knockdown (KD) tumour, despite not reaching statistical significance. The RNA expression of ICAM1 and VCAM1 genes also increased in the PAK4KD group [86]. Leukocyte transmigration and endothelial permeability are dependent on ICAM-1 expression [87,88,89]. Upon stimulation with cytokines, VCAM-1 is released from the endothelium and binds to integrins α4β1 and α4β7 on leukocytes. In addition to being involved in leukocyte adhesion and rolling, VCAM-1 also plays a role in leukocyte extravasation to some extent; however, its role in regulating vascular permeability has not been clarified [88,90]. In melanoma, Cercam1, Enpep, Itga3, and Lgals3 expressions were higher in tumours with PAK4 inhibition, indicating changes in angiogenesis [91].
The roles of PAKs in tumour vasculature are summarised in Table 2. Vascular changes will have a substantial impact on the effectiveness of chemotherapy and the infiltration of immune cells into the tumour. In the subsequent discussion, we will provide a summary of research findings that highlight the significance of PAKs inhibition in relation to chemotherapy responses and the activation of immune cells.

5. PAK and Therapeutical Responses

In pancreatic cancer, the extensive desmoplastic reaction leads to the formation of a dense stroma, causing inadequate vascularisation, inefficient drug delivery, and ineffective immune cell infiltration into the tumour sites, which eventually leads to therapeutical resistance and cancer progression [93]. PAKs have been recognised for their roles in the regulation of vasculature and therapeutical response. Here, we will discuss the impact of PAK effects on tumour vasculature in chemotherapy and immune response with a focus on pancreatic cancer.

5.1. PAK and Chemotherapy

PAKs are activated and play a crucial role in facilitating the resistance of cancer cells [94,95]. Modulating PAK activity holds the promise of sensitising cancer cells to chemotherapeutic agents, ultimately leading to an improved response to treatment. Shikonin, a natural inhibitor of PAK1, sensitises the pancreatic cancer cells to the chemotherapeutic drugs of gemcitabine and 5-FU [96]. A PAK1 inhibitor, FRAX597, when combined with gemcitabine, led to a synergistic inhibition of pancreatic cancer proliferation in vitro and a further reduced tumour growth in vivo [97]. The structural normalisation of blood vessels through PAK4 inhibition can enhance drug distribution and mitigate intra-tumoral hypoxia, ultimately resulting in enhanced tumour responses to molecular targeted therapy as well as radiotherapy and chemotherapy [85]. Inhibition of PAK4 not only reduces tumour growth but also enhances the efficacy of gemcitabine in mice carrying pancreatic cancer [98]. The combination of the PAK4 inhibitor, KPT-9274, and NAMPT modulators (KPT-9307) more effectively suppressed tumour growth in a xenografted mouse model [99]. The effects of PAK inhibition on the chemotherapy response of cancer cells are summarised in Table 3.

5.2. PAK in Tumour Immune Responses and Immunotherapy

Tumour vasculature normalisation would potentially facilitate immune cell infiltration and increase the efficacy of immunotherapy. Immune checkpoint inhibitors have been found to enhance vessel normalisation by engaging CD4+ T lymphocytes [100]. PD-L1 expression is upregulated via the activation of the PI3K/AKT and interferon-γ pathways, both of which have close interactions with PAK1 [59,101,102]. We have reported that PAK1 inhibition increased levels of intra-tumoral CD3+, CD4+, and CD8+ T cells and decreased PD-L1 expression of cancer cells, thereby stimulating anti-tumour immunity [81]. The observed diminished protein expression levels of αSMA and Desmin in the PAK1KO tumours suggest a potential impact on the deactivation of cancer-associated fibroblasts, which in turn may lead to the anticipated reduction in angiogenesis within the PAK1KO tumours [4,81].
PAK4 expression is negatively correlated with immune cell infiltration in other cancers. High PAK4 expression is associated with limited infiltration of T cells and dendritic cells in immune checkpoint inhibitor-resistant melanoma patients. However, genetic deletion of PAK4 reversed PD-1 blockade resistance by increasing CD8 T cell infiltration in mice carrying flank tumours of melanoma. A combination of anti-PD-1 treatment and a PAK4 inhibitor enhanced anti-tumour responses compared to anti-PD-1 treatment alone. The effects of PAK4 KO were mediated via inhibition of the WNT pathway [103]. Wnt/β-catenin signalling is involved in fundamental vascularisation processes, including sprouting and non-sprouting angiogenesis, vasculogenic mimicry, and the formation of mosaic vessels [104].
Inhibition of PAK4 enhanced the infiltration of CD103+ dendritic cells and CD8 T cells, and its expression correlates with CCL21 levels in melanoma biopsies. Notably, PAK4 KO cells exhibited increased expression of genes associated with blood vessel formation, including Cercam, Enpep, Itga, and Lgals. CD31, a primary blood vessel marker, was increased in tumours treated with anti-PD-1 in PAK4 KO mice [91]. In glioblastoma (GBM), inhibition of PAK4 has induced intra-tumoral CD3+ T-cell infiltration, rendering GBM more susceptible to the immunotherapy of CAR-T cells. It has been demonstrated that PAK4 inhibition transformed the disordered morphology of the tumour-associated vasculature, characterised by tortuous and disjointed vessels with spatial heterogeneity, into a well-organised structure marked by continuous vessels [85]. A similar effect has been observed in prostate cancer, where targeting PAK4 increases the infiltration of CD8 T cells and B cells into the tumour, reversing PD-1 blockade resistance. Inhibiting PAK4 also leads to increased angiogenic activities and increased expression of adhesion molecules in the TME, attracting CD8+ lymphocytes [86]. The effects of PAK1 and PAK4 on the tumour infiltration of immune cells and the associated changes in tumour vasculature have been summarised in Table 4.

6. Conclusions

Tumour vasculature plays a significant role in regulating the supply of nutrients, oxygen, and immune cell infiltration in the TME. Efforts in anti-angiogenesis treatments, designed to deprive cancer cells of nourishment by diminishing tumour blood vessel formation, have yielded outcomes below optimistic projection. The reduction in tumour vascularisation results in increased tumour hypoxia, leading to increased therapeutical resistance and cancer metastasis. The vasculature normalisation approach aims to reinstate proper blood flow, thereby improving the distribution of therapeutic drugs, enhancing the tumour infiltration of immune cells, and decreasing the spread of metastasis [105], which has important implications in pancreatic cancer where anti-angiogenesis treatment has limited effect.
In view of the importance of the immune system in cancer, it is crucial to understand the molecular mechanisms involved in immune cell infiltration and vasculature changes. The interaction between vasculature, tumour immune response, and PAKs in cancer biology presents a complicated and dynamic landscape. As simplified in Figure 4, the advancement in further exploring how PAKs affect tumour vasculature and change the tumour immune response will stimulate the development of novel strategies for the treatment of pancreatic cancer to improve patients’ survival.

Author Contributions

A.A. played a pivotal role in the conceptual design of the study, conducted an extensive literature review, and took the lead in drafting the manuscript. M.N. provided valuable input through a comprehensive review and played a crucial role in the finalisation of the manuscript. H.H. contributed significantly to the conceptual design of the study, made changes, and finalised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Pancare Foundation, Austin Medical Research Foundation (H.H.-2023, M.N.-2022), and MDHS (Medicine Dental Health Science, University of Melbourne) Seeding Ideas Grants (H.H., CA 2021).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

Arian is supported by the Melbourne Research Scholarship (MRS). Hong He is supported by the Henry Baldwin Cancer Research Trust Fund. All figures are created with BioRender.com.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

PAKsP21-activated kinases
PDAPancreatic ductal adenocarcinoma
TMETumour microenvironment
CSCsCancer stem cells
5-FU5-fluorouracil
TKIsTyrosine kinase inhibitors
PFSProgression-free survival
OSOverall survival
MVDMicrovessel density
GTPGuanosine triphosphate
MVIMicrovessel integrity
SEMA3ASemaphorin 3A
SalBSalvianolic acid B
HRGHistidine-rich glycoprotein
RGS5G-Protein Signalling 5
CdaCytidine deaminase
EMTEpithelial–mesenchymal transition
VSMCsVascular smooth muscle cells
CSF2Colony stimulating factor 2
HRGHeregulin β1
HIF-1αHypoxia-inducible factor 1 alpha
JMJD6Jumonji domain-containing protein 6
MMP9Matrix metalloproteinases 9
CAFsCancer-associated fibroblasts
PSCsPancreatic stellate cells
KOKnockout
KDKnockdown
GBM ECsGlioblastoma multiforme endothelial cells
IHCImmunohistochemical

References

  1. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef] [PubMed]
  2. Moletta, L.; Serafini, S.; Valmasoni, M.; Pierobon, E.S.; Ponzoni, A.; Sperti, C. Surgery for recurrent pancreatic cancer: Is it effective? Cancers 2019, 11, 991. [Google Scholar] [CrossRef] [PubMed]
  3. Adamska, A.; Domenichini, A.; Falasca, M. Pancreatic ductal adenocarcinoma: Current and evolving therapies. Int. J. Mol. Sci. 2017, 18, 1338. [Google Scholar] [CrossRef] [PubMed]
  4. Ansardamavandi, A.; Tafazzoli-Shadpour, M. The functional cross talk between cancer cells and cancer associated fibroblasts from a cancer mechanics perspective. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2021, 1868, 119103. [Google Scholar] [CrossRef] [PubMed]
  5. Krishna Priya, S.; Nagare, R.; Sneha, V.; Sidhanth, C.; Bindhya, S.; Manasa, P.; Ganesan, T. Tumour angiogenesis—Origin of blood vessels. Int. J. Cancer 2016, 139, 729–735. [Google Scholar] [CrossRef] [PubMed]
  6. Schaaf, M.B.; Garg, A.D.; Agostinis, P. Defining the role of the tumor vasculature in antitumor immunity and immunotherapy. Cell Death Dis. 2018, 9, 115. [Google Scholar] [CrossRef] [PubMed]
  7. Giantonio, B.J.; Catalano, P.J.; Meropol, N.J.; O’Dwyer, P.J.; Mitchell, E.P.; Alberts, S.R.; Schwartz, M.A.; Benson III, A.B. Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: Results from the Eastern Cooperative Oncology Group Study E3200. J. Clin. Oncol. 2007, 25, 1539–1544. [Google Scholar] [CrossRef]
  8. Cohen, M.H.; Shen, Y.L.; Keegan, P.; Pazdur, R. FDA drug approval summary: Bevacizumab (Avastin®) as treatment of recurrent glioblastoma multiforme. Oncologist 2009, 14, 1131–1138. [Google Scholar] [CrossRef]
  9. Heath, V.L.; Bicknell, R. Anticancer strategies involving the vasculature. Nat. Rev. Clin. Oncol. 2009, 6, 395–404. [Google Scholar] [CrossRef]
  10. Ivy, S.P.; Wick, J.Y.; Kaufman, B.M. An overview of small-molecule inhibitors of VEGFR signaling. Nat. Rev. Clin. Oncol. 2009, 6, 569–579. [Google Scholar] [CrossRef]
  11. Fudalej, M.; Kwaśniewska, D.; Nurzyński, P.; Badowska-Kozakiewicz, A.; Mękal, D.; Czerw, A.; Sygit, K.; Deptała, A. New Treatment Options in Metastatic Pancreatic Cancer. Cancers 2023, 15, 2327. [Google Scholar] [CrossRef] [PubMed]
  12. Van Cutsem, E.; Vervenne, W.L.; Bennouna, J.; Humblet, Y.; Gill, S.; Van Laethem, J.-L.; Verslype, C.; Scheithauer, W.; Shang, A.; Cosaert, J. Phase III trial of bevacizumab in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer. J. Clin. Oncol. 2009, 27, 2231–2237. [Google Scholar] [CrossRef] [PubMed]
  13. Vasudev, N.S.; Reynolds, A.R. Anti-angiogenic therapy for cancer: Current progress, unresolved questions and future directions. Angiogenesis 2014, 17, 471–494. [Google Scholar] [CrossRef] [PubMed]
  14. Azam, F.; Mehta, S.; Harris, A.L. Mechanisms of resistance to antiangiogenesis therapy. Eur. J. Cancer 2010, 46, 1323–1332. [Google Scholar] [CrossRef] [PubMed]
  15. Munshi, N.C.; Wilson, C. Increased bone marrow microvessel density in newly diagnosed multiple myeloma carries a poor prognosis. In Seminars in Oncology; Elsevier: Amsterdam, The Netherlands, 2001; pp. 565–569. [Google Scholar]
  16. Nishida, T.; Yoshitomi, H.; Takano, S.; Kagawa, S.; Shimizu, H.; Ohtsuka, M.; Kato, A.; Furukawa, K.; Miyazaki, M. Low stromal area and high stromal microvessel density predict poor prognosis in pancreatic cancer. Pancreas 2016, 45, 593–600. [Google Scholar] [CrossRef] [PubMed]
  17. van der Zee, J.A.; van Eijck, C.H.; Hop, W.C.; van Dekken, H.; Dicheva, B.M.; Seynhaeve, A.L.; Koning, G.A.; Eggermont, A.M.; ten Hagen, T.L. Angiogenesis: A prognostic determinant in pancreatic cancer? Eur. J. Cancer 2011, 47, 2576–2584. [Google Scholar] [CrossRef]
  18. MacLennan, G.T.; Bostwick, D.G. Microvessel density in renal cell carcinoma: Lack of prognostic significance. Urology 1995, 46, 27–30. [Google Scholar] [CrossRef] [PubMed]
  19. Carmeliet, P.; Jain, R.K. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat. Rev. Drug Discov. 2011, 10, 417–427. [Google Scholar] [CrossRef]
  20. Wang, S.; Zheng, Y.; Yang, F.; Zhu, L.; Zhu, X.-Q.; Wang, Z.-F.; Wu, X.-L.; Zhou, C.-H.; Yan, J.-Y.; Hu, B.-Y. The molecular biology of pancreatic adenocarcinoma: Translational challenges and clinical perspectives. Signal Transduct. Target. Ther. 2021, 6, 249. [Google Scholar] [CrossRef]
  21. Maitra, A.; Hruban, R.H. Pancreatic cancer. Annu. Rev. Pathol. Mech. Dis. 2008, 3, 157–188. [Google Scholar] [CrossRef]
  22. Yeo, D.; He, H.; Baldwin, G.S.; Nikfarjam, M. The role of p21-activated kinases in pancreatic cancer. Pancreas 2015, 44, 363–369. [Google Scholar] [CrossRef] [PubMed]
  23. Senapedis, W.; Crochiere, M.; Baloglu, E.; Landesman, Y. Therapeutic potential of targeting PAK signaling. Anti-Cancer Agents Med. Chem. (Former. Curr. Med. Chem.-Anti-Cancer Agents) 2016, 16, 75–88. [Google Scholar] [CrossRef] [PubMed]
  24. Baluk, P.; Hashizume, H.; McDonald, D.M. Cellular abnormalities of blood vessels as targets in cancer. Curr. Opin. Genet. Dev. 2005, 15, 102–111. [Google Scholar] [CrossRef] [PubMed]
  25. Helmlinger, G.; Yuan, F.; Dellian, M.; Jain, R.K. Interstitial pH and pO2 gradients in solid tumors in vivo: High-resolution measurements reveal a lack of correlation. Nat. Med. 1997, 3, 177–182. [Google Scholar] [CrossRef] [PubMed]
  26. Duda, D.G.; Jain, R.K.; Willett, C.G. Antiangiogenics: The potential role of integrating this novel treatment modality with chemoradiation for solid cancers. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2007, 25, 4033. [Google Scholar] [CrossRef] [PubMed]
  27. Pouysségur, J.; Dayan, F.; Mazure, N.M. Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature 2006, 441, 437–443. [Google Scholar] [CrossRef] [PubMed]
  28. Bansod, S.; Dodhiawala, P.B.; Lim, K.-H. Oncogenic KRAS-induced feedback inflammatory signaling in pancreatic cancer: An overview and new therapeutic opportunities. Cancers 2021, 13, 5481. [Google Scholar] [CrossRef]
  29. Erkan, M.; Hausmann, S.; Michalski, C.W.; Fingerle, A.A.; Dobritz, M.; Kleeff, J.; Friess, H. The role of stroma in pancreatic cancer: Diagnostic and therapeutic implications. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 454–467. [Google Scholar] [CrossRef]
  30. Paez-Ribes, M.; Allen, E.; Hudock, J.; Takeda, T.; Okuyama, H.; Viñals, F.; Inoue, M.; Bergers, G.; Hanahan, D.; Casanovas, O. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 2009, 15, 220–231. [Google Scholar] [CrossRef]
  31. Shao, Y.Y.; Lu, L.C.; Cheng, A.L.; Hsu, C.H. Increasing incidence of brain metastasis in patients with advanced hepatocellular carcinoma in the era of antiangiogenic targeted therapy. Oncologist 2011, 16, 82–86. [Google Scholar] [CrossRef]
  32. Zhu, X.-D.; Sun, H.-C.; Xu, H.-X.; Kong, L.-Q.; Chai, Z.-T.; Lu, L.; Zhang, J.-B.; Gao, D.-M.; Wang, W.-Q.; Zhang, W. Antiangiogenic therapy promoted metastasis of hepatocellular carcinoma by suppressing host-derived interleukin-12b in mouse models. Angiogenesis 2013, 16, 809–820. [Google Scholar] [CrossRef] [PubMed]
  33. Yin, T.; He, S.; Ye, T.; Shen, G.; Wan, Y.; Wang, Y. Antiangiogenic therapy using sunitinib combined with rapamycin retards tumor growth but promotes metastasis. Transl. Oncol. 2014, 7, 221–229. [Google Scholar] [CrossRef] [PubMed]
  34. Li, S.; Xu, H.-X.; Wu, C.-T.; Wang, W.-Q.; Jin, W.; Gao, H.-L.; Li, H.; Zhang, S.-R.; Xu, J.-Z.; Qi, Z.-H. Angiogenesis in pancreatic cancer: Current research status and clinical implications. Angiogenesis 2019, 22, 15–36. [Google Scholar] [CrossRef] [PubMed]
  35. Bergers, G.; Hanahan, D. Modes of resistance to anti-angiogenic therapy. Nat. Rev. Cancer 2008, 8, 592–603. [Google Scholar] [CrossRef] [PubMed]
  36. Viallard, C.; Larrivée, B. Tumor angiogenesis and vascular normalization: Alternative therapeutic targets. Angiogenesis 2017, 20, 409–426. [Google Scholar] [CrossRef] [PubMed]
  37. Abdalla, A.M.; Xiao, L.; Ullah, M.W.; Yu, M.; Ouyang, C.; Yang, G. Current challenges of cancer anti-angiogenic therapy and the promise of nanotherapeutics. Theranostics 2018, 8, 533. [Google Scholar] [CrossRef] [PubMed]
  38. Gilles, M.-E.; Maione, F.; Cossutta, M.; Carpentier, G.; Caruana, L.; Di Maria, S.; Houppe, C.; Destouches, D.; Shchors, K.; Prochasson, C. Nucleolin targeting impairs the progression of pancreatic cancer and promotes the normalization of tumor vasculature. Cancer Res. 2016, 76, 7181–7193. [Google Scholar] [CrossRef]
  39. Raggi, F.; Pelassa, S.; Pierobon, D.; Penco, F.; Gattorno, M.; Novelli, F.; Eva, A.; Varesio, L.; Giovarelli, M.; Bosco, M.C. Regulation of human macrophage M1–M2 polarization balance by hypoxia and the triggering receptor expressed on myeloid cells-1. Front. Immunol. 2017, 8, 1097. [Google Scholar] [CrossRef]
  40. Huang, Y.; Yuan, J.; Righi, E.; Kamoun, W.S.; Ancukiewicz, M.; Nezivar, J.; Santosuosso, M.; Martin, J.D.; Martin, M.R.; Vianello, F. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc. Natl. Acad. Sci. USA 2012, 109, 17561–17566. [Google Scholar] [CrossRef]
  41. Serini, G.; Valdembri, D.; Zanivan, S.; Morterra, G.; Burkhardt, C.; Caccavari, F.; Zammataro, L.; Primo, L.; Tamagnone, L.; Logan, M. Class 3 semaphorins control vascular morphogenesis by inhibiting integrin function. Nature 2003, 424, 391–397. [Google Scholar] [CrossRef]
  42. Nagathihalli, N.S.; Castellanos, J.A.; Shi, C.; Beesetty, Y.; Reyzer, M.L.; Caprioli, R.; Chen, X.; Walsh, A.J.; Skala, M.C.; Moses, H.L. Signal transducer and activator of transcription 3, mediated remodeling of the tumor microenvironment results in enhanced tumor drug delivery in a mouse model of pancreatic cancer. Gastroenterology 2015, 149, 1932–1943. [Google Scholar] [CrossRef] [PubMed]
  43. Qian, C.; Yang, C.; Tang, Y.; Zheng, W.; Zhou, Y.; Zhang, S.; Song, M.; Cheng, P.; Wei, Z.; Zhong, C. Pharmacological manipulation of Ezh2 with salvianolic acid B results in tumor vascular normalization and synergizes with cisplatin and T cell-mediated immunotherapy. Pharmacol. Res. 2022, 182, 106333. [Google Scholar] [CrossRef] [PubMed]
  44. Poon, I.K.; Patel, K.K.; Davis, D.S.; Parish, C.R.; Hulett, M.D. Histidine-rich glycoprotein: The Swiss Army knife of mammalian plasma. Blood J. Am. Soc. Hematol. 2011, 117, 2093–2101. [Google Scholar] [CrossRef] [PubMed]
  45. Rolny, C.; Mazzone, M.; Tugues, S.; Laoui, D.; Johansson, I.; Coulon, C.; Squadrito, M.L.; Segura, I.; Li, X.; Knevels, E. HRG inhibits tumor growth and metastasis by inducing macrophage polarization and vessel normalization through downregulation of PlGF. Cancer Cell 2011, 19, 31–44. [Google Scholar] [CrossRef] [PubMed]
  46. Silini, A.; Ghilardi, C.; Figini, S.; Sangalli, F.; Fruscio, R.; Dahse, R.; Pedley, R.B.; Giavazzi, R.; Bani, M. Regulator of G-protein signaling 5 (RGS5) protein: A novel marker of cancer vasculature elicited and sustained by the tumor’s proangiogenic microenvironment. Cell. Mol. Life Sci. 2012, 69, 1167–1178. [Google Scholar] [CrossRef] [PubMed]
  47. Hamzah, J.; Jugold, M.; Kiessling, F.; Rigby, P.; Manzur, M.; Marti, H.H.; Rabie, T.; Kaden, S.; Gröne, H.-J.; Hämmerling, G.J. Vascular normalization in Rgs5-deficient tumours promotes immune destruction. Nature 2008, 453, 410–414. [Google Scholar] [CrossRef] [PubMed]
  48. Stockton, R.A.; Schaefer, E.; Schwartz, M.A. p21-activated kinase regulates endothelial permeability through modulation of contractility. J. Biol. Chem. 2004, 279, 46621–46630. [Google Scholar] [CrossRef] [PubMed]
  49. Fryer, B.H.; Field, J. Rho, Rac, Pak and angiogenesis: Old roles and newly identified responsibilities in endothelial cells. Cancer Lett. 2005, 229, 13–23. [Google Scholar] [CrossRef]
  50. Wang, W.-Q.; Liu, L.; Sun, H.-C.; Fu, Y.-L.; Xu, H.-X.; Chai, Z.-T.; Zhang, Q.-B.; Kong, L.-Q.; Zhu, X.-D.; Lu, L. Tanshinone IIA inhibits metastasis after palliative resection of hepatocellular carcinoma and prolongs survival in part via vascular normalization. J. Hematol. Oncol. 2012, 5, 69. [Google Scholar] [CrossRef]
  51. Zhang, H.; Ren, Y.; Tang, X.; Wang, K.; Liu, Y.; Zhang, L.; Li, X.; Liu, P.; Zhao, C.; He, J. Vascular normalization induced by sinomenine hydrochloride results in suppressed mammary tumor growth and metastasis. Sci. Rep. 2015, 5, 8888. [Google Scholar] [CrossRef]
  52. Waters, A.M.; Der, C.J. KRAS: The critical driver and therapeutic target for pancreatic cancer. Cold Spring Harb. Perspect. Med. 2017, 8, a031435. [Google Scholar] [CrossRef] [PubMed]
  53. Manser, E.; Leung, T.; Salihuddin, H.; Zhao, Z.-s.; Lim, L. A brain serine/threonine protein kinase activated by Cdc42 and Rac1. Nature 1994, 367, 40–46. [Google Scholar] [CrossRef] [PubMed]
  54. Ma, Y.; Nikfarjam, M.; He, H. The trilogy of P21 activated kinase, autophagy and immune evasion in pancreatic ductal adenocarcinoma. Cancer Lett. 2022, 548, 215868. [Google Scholar] [CrossRef] [PubMed]
  55. Ahn, A.R.; Karamikheirabad, M.; Park, M.S.; Zhang, J.; Kim, H.S.; Jeong, J.S.; Kim, K.M.; Park, H.S.; Jang, K.Y. Expression Patterns of PAK4 and PHF8 Are Associated with the Survival of Gallbladder Carcinoma Patients. Diagnostics 2023, 13, 1149. [Google Scholar] [CrossRef] [PubMed]
  56. Tang, L.; Gao, Y.; Li, T. Pan-cancer analysis identifies the immunological and prognostic role of PAK4. Life Sci. 2023, 312, 121263. [Google Scholar] [CrossRef] [PubMed]
  57. Park, J.; Kim, J.m.; Park, J.K.; Huang, S.; Kwak, S.Y.; Ryu, K.A.; Kong, G.; Park, J.; Koo, B.S. Association of p21-activated kinase-1 activity with aggressive tumor behavior and poor prognosis of head and neck cancer. Head Neck 2015, 37, 953–963. [Google Scholar] [CrossRef] [PubMed]
  58. Jagadeeshan, S.; Krishnamoorthy, Y.; Singhal, M.; Subramanian, A.; Mavuluri, J.; Lakshmi, A.; Roshini, A.; Baskar, G.; Ravi, M.; Joseph, L. Transcriptional regulation of fibronectin by p21-activated kinase-1 modulates pancreatic tumorigenesis. Oncogene 2015, 34, 455–464. [Google Scholar] [CrossRef]
  59. Wang, K.; Baldwin, G.S.; Nikfarjam, M.; He, H. p21-activated kinase signalling in pancreatic cancer: New insights into tumour biology and immune modulation. World J. Gastroenterol. 2018, 24, 3709. [Google Scholar] [CrossRef]
  60. Hoff, P.M.; Machado, K.K. Role of angiogenesis in the pathogenesis of cancer. Cancer Treat. Rev. 2012, 38, 825–833. [Google Scholar] [CrossRef]
  61. Yuan, L.; Duan, X.; Dong, J.; Lu, Q.; Zhou, J.; Zhao, Z.; Bao, J.; Jing, Z. p21-activated kinase 4 promotes intimal hyperplasia and vascular smooth muscle cells proliferation during superficial femoral artery restenosis after angioplasty. BioMed Res. Int. 2017, 2017, 5296516. [Google Scholar] [CrossRef]
  62. Rane, C.K.; Minden, A. P21 activated kinase signaling in cancer. In Seminars in Cancer Biology; Academic Press: Cambridge, MA, USA, 2019; pp. 40–49. [Google Scholar]
  63. King, H.; Thillai, K.; Whale, A.; Arumugam, P.; Eldaly, H.; Kocher, H.M.; Wells, C.M. PAK4 interacts with p85 alpha: Implications for pancreatic cancer cell migration. Sci. Rep. 2017, 7, 42575. [Google Scholar] [CrossRef] [PubMed]
  64. Thillai, K.; Sarker, D.; Wells, C. PAK4 as a potential therapeutic target in pancreatic ductal adenocarcinoma (PDAC). J. Clin. Oncol. 2017, 35, e23139. [Google Scholar] [CrossRef]
  65. Tyagi, N.; Marimuthu, S.; Bhardwaj, A.; Deshmukh, S.K.; Srivastava, S.K.; Singh, A.P.; McClellan, S.; Carter, J.E.; Singh, S. p-21 activated kinase 4 (PAK4) maintains stem cell-like phenotypes in pancreatic cancer cells through activation of STAT3 signaling. Cancer Lett. 2016, 370, 260–267. [Google Scholar] [CrossRef] [PubMed]
  66. Wang, S.; Wang, S.-Y.; Du, F.; Han, Q.; Wang, E.-H.; Luo, E.-J.; Liu, Y. Knockdown of PAK1 inhibits the proliferation and invasion of Non–Small cell Lung cancer cells through the ERK pathway. Appl. Immunohistochem. Mol. Morphol. 2020, 28, 602–610. [Google Scholar] [CrossRef] [PubMed]
  67. Davidson, A.; Tyler, J.; Hume, P.; Singh, V.; Koronakis, V. A kinase-independent function of PAK is crucial for pathogen-mediated actin remodelling. PLoS Pathog. 2021, 17, e1009902. [Google Scholar] [CrossRef]
  68. Li, Z.; Zhang, H.; Lundin, L.; Thullberg, M.; Liu, Y.; Wang, Y.; Claesson-Welsh, L.; Strömblad, S. p21-activated kinase 4 phosphorylation of integrin β5 Ser-759 and Ser-762 regulates cell migration. J. Biol. Chem. 2010, 285, 23699–23710. [Google Scholar] [CrossRef] [PubMed]
  69. Yi, K.W.; Kim, S.H.; Ihm, H.J.; Oh, Y.S.; Chae, H.D.; Kim, C.-H.; Kang, B.M. Increased expression of p21-activated kinase 4 in adenomyosis and its regulation of matrix metalloproteinase-2 and-9 in endometrial cells. Fertil. Steril. 2015, 103, 1089–1097.e1082. [Google Scholar] [CrossRef]
  70. Etienne-Manneville, S.; Hall, A. Rho GTPases in cell biology. Nature 2002, 420, 629–635. [Google Scholar] [CrossRef]
  71. Bryan, B.A.; Dennstedt, E.; Mitchell, D.C.; Walshe, T.E.; Noma, K.; Loureiro, R.; Saint-Geniez, M.; Campaigniac, J.-P.; Liao, J.K.; D’Amore, P.A. RhoA/ROCK signaling is essential for multiple aspects of VEGF-mediated angiogenesis. FASEB J. 2010, 24, 3186. [Google Scholar] [CrossRef]
  72. Bryan, B.; D’amore, P. What tangled webs they weave: Rho-GTPase control of angiogenesis. Cell. Mol. Life Sci. 2007, 64, 2053–2065. [Google Scholar] [CrossRef]
  73. Hu, G.-D.; Chen, Y.-H.; Zhang, L.; Tong, W.-C.; Cheng, Y.-X.; Luo, Y.-L.; Cai, S.-X.; Zhang, L. The generation of the endothelial specific cdc42-deficient mice and the effect of cdc42 deletion on the angiogenesis and embryonic development. Chin. Med. J. 2011, 124, 4155–4159. [Google Scholar]
  74. El Atat, O.; Fakih, A.; El-Sibai, M. RHOG activates RAC1 through CDC42 leading to tube formation in vascular endothelial cells. Cells 2019, 8, 171. [Google Scholar] [CrossRef]
  75. Li, C.-F.; Chan, T.-C.; Fang, F.-M.; Yu, S.-C.; Huang, H.-Y. PAK1 overexpression promotes myxofibrosarcoma angiogenesis through STAT5B-mediated CSF2 transactivation: Clinical and therapeutic relevance of amplification and nuclear entry. Int. J. Biol. Sci. 2023, 19, 3920. [Google Scholar] [CrossRef] [PubMed]
  76. Bagheri-Yarmand, R.; Vadlamudi, R.K.; Wang, R.-A.; Mendelsohn, J.; Kumar, R. Vascular endothelial growth factor up-regulation via p21-activated kinase-1 signaling regulates heregulin-β1-mediated angiogenesis. J. Biol. Chem. 2000, 275, 39451–39457. [Google Scholar] [CrossRef] [PubMed]
  77. Jiang, S.; Gao, Y.; Yu, Q.H.; Li, M.; Cheng, X.; Hu, S.B.; Song, Z.F.; Zheng, Q.C. P-21-activated kinase 1 contributes to tumor angiogenesis upon photodynamic therapy via the HIF-1α/VEGF pathway. Biochem. Biophys. Res. Commun. 2020, 526, 98–104. [Google Scholar] [CrossRef] [PubMed]
  78. Liao, D.; Johnson, R.S. Hypoxia: A key regulator of angiogenesis in cancer. Cancer Metastasis Rev. 2007, 26, 281–290. [Google Scholar] [CrossRef]
  79. Goc, A.; Al-Azayzih, A.; Abdalla, M.; Al-Husein, B.; Kavuri, S.; Lee, J.; Moses, K.; Somanath, P.R. P21 activated kinase-1 (Pak1) promotes prostate tumor growth and microinvasion via inhibition of transforming growth factor β expression and enhanced matrix metalloproteinase 9 secretion. J. Biol. Chem. 2013, 288, 3025–3035. [Google Scholar] [CrossRef] [PubMed]
  80. Li, T.; Li, Y.-G.; Pu, D.-M. Matrix metalloproteinase-2 and-9 expression correlated with angiogenesis in human adenomyosis. Gynecol. Obstet. Investig. 2006, 62, 229–235. [Google Scholar] [CrossRef] [PubMed]
  81. Wang, K.; Zhan, Y.; Huynh, N.; Dumesny, C.; Wang, X.; Asadi, K.; Herrmann, D.; Timpson, P.; Yang, Y.; Walsh, K. Inhibition of PAK1 suppresses pancreatic cancer by stimulation of anti-tumour immunity through down-regulation of PD-L1. Cancer Lett. 2020, 472, 8–18. [Google Scholar] [CrossRef]
  82. Yeo, D.; Phillips, P.; Baldwin, G.S.; He, H.; Nikfarjam, M. Inhibition of group 1 p21-activated kinases suppresses pancreatic stellate cell activation and increases survival of mice with pancreatic cancer. Int. J. Cancer 2017, 140, 2101–2111. [Google Scholar] [CrossRef]
  83. Allam, A.; Thomsen, A.; Gothwal, M.; Saha, D.; Maurer, J.; Brunner, T. Pancreatic stellate cells in pancreatic cancer: In focus. Pancreatology 2017, 17, 514–522. [Google Scholar] [CrossRef] [PubMed]
  84. Topalovski, M.; Brekken, R.A. Matrix control of pancreatic cancer: New insights into fibronectin signaling. Cancer Lett. 2016, 381, 252–258. [Google Scholar] [CrossRef] [PubMed]
  85. Ma, W.; Wang, Y.; Zhang, R.; Yang, F.; Zhang, D.; Huang, M.; Zhang, L.; Dorsey, J.F.; Binder, Z.A.; O’Rourke, D.M. Targeting PAK4 to reprogram the vascular microenvironment and improve CAR-T immunotherapy for glioblastoma. Nat. Cancer 2021, 2, 83–97. [Google Scholar] [CrossRef]
  86. Su, S.; You, S.; Wang, Y.; Tamukong, P.; Quist, M.J.; Grasso, C.S.; Kim, H.L. PAK4 inhibition improves PD1 blockade immunotherapy in prostate cancer by increasing immune infiltration. Cancer Lett. 2023, 555, 216034. [Google Scholar] [CrossRef] [PubMed]
  87. Frank, P.G.; Lisanti, M.P. ICAM-1: Role in inflammation and in the regulation of vascular permeability. Am. J. Physiol. -Heart Circ. Physiol. 2008, 295, H926–H927. [Google Scholar] [CrossRef]
  88. Sarelius, I.H.; Glading, A.J. Control of vascular permeability by adhesion molecules. Tissue Barriers 2015, 3, e985954. [Google Scholar] [CrossRef] [PubMed]
  89. Bui, T.M.; Wiesolek, H.L.; Sumagin, R. ICAM-1: A master regulator of cellular responses in inflammation, injury resolution, and tumorigenesis. J. Leucoc. Biol. 2020, 108, 787–799. [Google Scholar] [CrossRef]
  90. Riegler, J.; Gill, H.; Ogasawara, A.; Hedehus, M.; Javinal, V.; Oeh, J.; Ferl, G.Z.; Marik, J.; Williams, S.; Sampath, D. VCAM-1 density and tumor perfusion predict T-cell infiltration and treatment response in preclinical models. Neoplasia 2019, 21, 1036–1050. [Google Scholar] [CrossRef]
  91. Abril-Rodriguez, G.; Torrejon, D.Y.; Karin, D.; Campbell, K.M.; Medina, E.; Saco, J.D.; Galvez, M.; Champhekar, A.S.; Perez-Garcilazo, I.; Baselga-Carretero, I. Remodeling of the tumor microenvironment through PAK4 inhibition sensitizes tumors to immune checkpoint blockade. Cancer Res. Commun. 2022, 2, 1214–1228. [Google Scholar] [CrossRef]
  92. Liu, X.; Si, W.; Liu, X.; He, L.; Ren, J.; Yang, Z.; Yang, J.; Li, W.; Liu, S.; Pei, F. JMJD6 promotes melanoma carcinogenesis through regulation of the alternative splicing of PAK1, a key MAPK signaling component. Mol. Cancer 2017, 16, 1–18. [Google Scholar] [CrossRef]
  93. Neesse, A.; Michl, P.; Frese, K.K.; Feig, C.; Cook, N.; Jacobetz, M.A.; Lolkema, M.P.; Buchholz, M.; Olive, K.P.; Gress, T.M. Stromal biology and therapy in pancreatic cancer. Gut 2011, 60, 861–868. [Google Scholar] [CrossRef] [PubMed]
  94. Martin, K.; Pritchett, J.; Llewellyn, J.; Mullan, A.F.; Athwal, V.S.; Dobie, R.; Harvey, E.; Zeef, L.; Farrow, S.; Streuli, C. PAK proteins and YAP-1 signalling downstream of integrin beta-1 in myofibroblasts promote liver fibrosis. Nat. Commun. 2016, 7, 12502. [Google Scholar] [CrossRef] [PubMed]
  95. Lu, H.; Liu, S.; Zhang, G.; Wu, B.; Zhu, Y.; Frederick, D.T.; Hu, Y.; Zhong, W.; Randell, S.; Sadek, N. PAK signalling drives acquired drug resistance to MAPK inhibitors in BRAF-mutant melanomas. Nature 2017, 550, 133–136. [Google Scholar] [CrossRef] [PubMed]
  96. Ji, W.; Sun, X.; Gao, Y.; Lu, M.; Zhu, L.; Wang, D.; Hu, C.; Chen, J.; Cao, P. Natural Compound Shikonin Is a Novel PAK1 Inhibitor and Enhances Efficacy of Chemotherapy against Pancreatic Cancer Cells. Molecules 2022, 27, 2747. [Google Scholar] [CrossRef] [PubMed]
  97. Yeo, D.; He, H.; Patel, O.; Lowy, A.M.; Baldwin, G.S.; Nikfarjam, M. FRAX597, a PAK1 inhibitor, synergistically reduces pancreatic cancer growth when combined with gemcitabine. BMC Cancer 2016, 16, 24. [Google Scholar] [CrossRef] [PubMed]
  98. He, H.; Dumesny, C.; Ang, C.-S.; Dong, L.; Ma, Y.; Zeng, J.; Nikfarjam, M. A novel PAK4 inhibitor suppresses pancreatic cancer growth and enhances the inhibitory effect of gemcitabine. Transl. Oncol. 2022, 16, 101329. [Google Scholar] [CrossRef] [PubMed]
  99. Mohammad, R.M.; Li, Y.; Muqbil, I.; Aboukameel, A.; Senapedis, W.; Baloglu, E.; Landesman, Y.; Philip, P.A.; Azmi, A.S. Targeting Rho GTPase effector p21 activated kinase 4 (PAK4) suppresses p-Bad-microRNA drug resistance axis leading to inhibition of pancreatic ductal adenocarcinoma proliferation. Small GTPases 2019, 10, 367–377. [Google Scholar] [CrossRef]
  100. Tian, L.; Goldstein, A.; Wang, H.; Ching Lo, H.; Sun Kim, I.; Welte, T.; Sheng, K.; Dobrolecki, L.E.; Zhang, X.; Putluri, N. Mutual regulation of tumour vessel normalization and immunostimulatory reprogramming. Nature 2017, 544, 250–254. [Google Scholar] [CrossRef]
  101. Parsa, A.T.; Waldron, J.S.; Panner, A.; Crane, C.A.; Parney, I.F.; Barry, J.J.; Cachola, K.E.; Murray, J.C.; Tihan, T.; Jensen, M.C. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat. Med. 2007, 13, 84–88. [Google Scholar] [CrossRef]
  102. Kharma, B.; Baba, T.; Matsumura, N.; Kang, H.S.; Hamanishi, J.; Murakami, R.; McConechy, M.M.; Leung, S.; Yamaguchi, K.; Hosoe, Y. STAT1 drives tumor progression in serous papillary endometrial cancer. Cancer Res. 2014, 74, 6519–6530. [Google Scholar] [CrossRef]
  103. Abril-Rodriguez, G.; Torrejon, D.Y.; Liu, W.; Zaretsky, J.M.; Nowicki, T.S.; Tsoi, J.; Puig-Saus, C.; Baselga-Carretero, I.; Medina, E.; Quist, M.J. PAK4 inhibition improves PD-1 blockade immunotherapy. Nat. Cancer 2020, 1, 46–58. [Google Scholar] [CrossRef]
  104. Kasprzak, A. Angiogenesis-related functions of Wnt signaling in colorectal carcinogenesis. Cancers 2020, 12, 3601. [Google Scholar] [CrossRef]
  105. Cully, M. Tumour vessel normalization takes centre stage. Nat. Rev. Drug Discov. 2017, 16, 87. [Google Scholar] [CrossRef]
Figure 1. Factors contributing to abnormal tumour vasculature. One of the essential components within the tumour microenvironment (TME) is the existence of abnormal vascular structures. Abnormal blood vessel growth, leaky vessel, loss of pericyte support, and low levels of oxygen are primary characteristics of these blood vessels. These features are interconnected and can mutually enhance the abnormalisation of blood vessels. For instance, a hypoxic microenvironment triggers the production of VEGF, which in turn reinforces the growth of abnormal blood vessels [27].
Figure 1. Factors contributing to abnormal tumour vasculature. One of the essential components within the tumour microenvironment (TME) is the existence of abnormal vascular structures. Abnormal blood vessel growth, leaky vessel, loss of pericyte support, and low levels of oxygen are primary characteristics of these blood vessels. These features are interconnected and can mutually enhance the abnormalisation of blood vessels. For instance, a hypoxic microenvironment triggers the production of VEGF, which in turn reinforces the growth of abnormal blood vessels [27].
Cells 12 02692 g001
Figure 2. Anti-angiogenesis vs. vascular normalisation. The conventional treatment of cancer with anti-angiogenesis drugs is associated with negative outcomes. Despite limiting the amount of nutrition delivered to cancer cells, it can also increase hypoxia, resulting in increased metastasis. Drug delivery to the tumour site can also be restricted by anti-angiogenesis. Alternatively, vascular normalisation can reduce levels of hypoxia and enhance the tumour infiltration of immune cells and drug delivery to the tumour site. To normalise tumour vasculature and limit tumour growth, restoring pericyte support is essential. It has been demonstrated in PDA models that increasing pericyte coverage can reduce hypoxia and increase drug delivery [38].
Figure 2. Anti-angiogenesis vs. vascular normalisation. The conventional treatment of cancer with anti-angiogenesis drugs is associated with negative outcomes. Despite limiting the amount of nutrition delivered to cancer cells, it can also increase hypoxia, resulting in increased metastasis. Drug delivery to the tumour site can also be restricted by anti-angiogenesis. Alternatively, vascular normalisation can reduce levels of hypoxia and enhance the tumour infiltration of immune cells and drug delivery to the tumour site. To normalise tumour vasculature and limit tumour growth, restoring pericyte support is essential. It has been demonstrated in PDA models that increasing pericyte coverage can reduce hypoxia and increase drug delivery [38].
Cells 12 02692 g002
Figure 3. Activation and functions of PAKs in pancreatic cancer. PAK1 and PAK4 act downstream of KRas, stimulate cell proliferation, epithelial–mesenchyme transition (EMT), and migration/invasion, and contribute to abnormal vasculature and immune suppression.
Figure 3. Activation and functions of PAKs in pancreatic cancer. PAK1 and PAK4 act downstream of KRas, stimulate cell proliferation, epithelial–mesenchyme transition (EMT), and migration/invasion, and contribute to abnormal vasculature and immune suppression.
Cells 12 02692 g003
Figure 4. Significance of the effects of PAKs on tumour vasculature in cancer treatment.
Figure 4. Significance of the effects of PAKs on tumour vasculature in cancer treatment.
Cells 12 02692 g004
Table 2. PAKs in tumour vasculature.
Table 2. PAKs in tumour vasculature.
Type of CancerTreatmentResultOtherRef.
Myxofibrosarcoma↑ PAK1↑ Tube formation and, ↑ Microvascular density (CD31 expression).↑ CSF2 expression.[75]
Breast↑ PAK1↑ Angiogenesis (CD34 expression).↑ VEGF.[76]
Cholangiocarcinoma↓ PAK1↓ Tube formation and angiogenesis. ↓ VEGF. [77]
Melanoma↓ PAK1↓ Tube formation and number of blood vessels.PAK1 expression is regulated by JMJD6, which phosphorylates RAF and MEK and positively regulates the MAPK pathway.[92]
Prostate↓ PAK1↓ Laminin positive blood vessels.↓ MMP9 (tenfold).[79]
Pancreatic ↓ PAK1↓ αSMA and Desmin.Deactivation of PSCs contributing to angiogenesis.[81,82]
Glioblastoma endothelial cells↓ PAK4↑ Tube formation.
≈Change of CD31 expression.
↓ Monolayer permeability.
↓ Hypoxia and vascular abnormality.
↓ Fibroblast-specific protein 1 (FSP-1), αsma, CDH2 (N-cadherin), SLUG, and ZEB1.
↑ Claudin-1, Claudin-14, ICAM1, and VCAM1 expression (twofold).
[85]
Prostate ↓ PAK4↑ Blood vessel width and density (PECAM1).↑ ICAM1 (CD54), VCAM1 (CD106), VEGFA RNA sequencing, and PECAM1 (CD31).[86]
Melanoma↓ PAK4↑ Cercam1, Enpep, Itga3, and Lgals3. [91]
Table 3. Inhibition of PAKs enhances cancer cells’ response to chemotherapy drugs.
Table 3. Inhibition of PAKs enhances cancer cells’ response to chemotherapy drugs.
Cancer TypePAK InhibitorsTargetTreatmentMechanismRef.
MelanomaPF-3758309PAK1
PAK4
BRAFi inhibitor
MEKi inhibitor
Enhance apoptosis and reduce cellular resistance to combined therapy via regulation of multiple pathways including JNK, ERK, β-catenin, and mTOR. [95]
PancreaticShikoninPAK1Gemcitabine
5-FU
Increase apoptosis in cancer cells.[96]
PancreaticFRAX597PAK1GemcitabineSuppress HIF1α and AKT.[97]
PancreaticPF-3758309PAK1
PAK4
Gemcitabine
5-FU
Abraxane
Reduce cell proliferation and enhance apoptosis.
Decrease the expression of αSMA, Phalloidin, and HIF1α in vivo.
[100]
PancreaticPAKibPAK4GemcitabineInduce cell cycle arrest and cell death and regulate cell junction and adhesion.[98]
PancreaticKPT-9274
KPT-9307
PAK4GemcitabineDownregulate p-Bad-microRNA drug resistance axis and upregulate tumour-suppressive miRNAs.[99]
Table 4. Inhibition of PAKs causes the infiltration of immune cells into the tumour area.
Table 4. Inhibition of PAKs causes the infiltration of immune cells into the tumour area.
Type of CancerTreatmentTechniquesType of Immune Cell ActivationPossible Effect on Tumour VasculatureRef.
Pancreatic ↓ PAK1Western blot and IHCT cells (CD3+, CD4+, and CD8+)↓ αSMA and Desmin, which indicates cancer associated fibroblast deactivation, in turn anticipating the reduced angiogenesis. [81]
Melanoma↓ PAK4Mass cytometry and IHCT cells (CD8+) and dendritic cells↑ Activation of WNT signalling pathway, anticipating the increasing angiogenesis.[103]
Melanoma↓ PAK4Transcriptomic, IHC, and flow cytometryT cells (CD8+) and dendritic cells (CD103+)↑ Alterations in genes associated with blood vessel formation, specifically, Cercam, Enpep, Itga, and Lgals.[91]
Glioblastoma↓ PAK4IHC and bioluminescence imagingT cells (CD3+)↑ Vascular normality with reduced hypoxia.[85]
Prostate↓ PAK4Flow cytometry and IHCT cells (CD8+) and B cells↑ ICAM1 and VCAM1.
↑ Inflammatory signals (CCR7, CCL19 and CCL21, CXCL13, and CXCR5).
[86]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ansardamavandi, A.; Nikfarjam, M.; He, H. PAK in Pancreatic Cancer-Associated Vasculature: Implications for Therapeutic Response. Cells 2023, 12, 2692. https://doi.org/10.3390/cells12232692

AMA Style

Ansardamavandi A, Nikfarjam M, He H. PAK in Pancreatic Cancer-Associated Vasculature: Implications for Therapeutic Response. Cells. 2023; 12(23):2692. https://doi.org/10.3390/cells12232692

Chicago/Turabian Style

Ansardamavandi, Arian, Mehrdad Nikfarjam, and Hong He. 2023. "PAK in Pancreatic Cancer-Associated Vasculature: Implications for Therapeutic Response" Cells 12, no. 23: 2692. https://doi.org/10.3390/cells12232692

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop