Elsevier

Bone

Volume 119, February 2019, Pages 82-86
Bone

Review Article
Parallels between hematopoietic stem cell and prostate cancer disseminated tumor cell regulation

https://doi.org/10.1016/j.bone.2018.02.025Get rights and content

Highlights

  • Prostate cancer disseminated tumor cells (DTCs) and hematopoietic stem cells (HSCs) have analogous functions.

  • Both home to the bone marrow, for which CXCL12 is critically important.

  • HSCs and DTCs share a home or “niche” as shown in part by competition experiments.

  • The niche regulates survival and dormancy vs. proliferation of both cell types.

Abstract

The bone marrow is the primary site of hematopoiesis and the home for hematopoietic stem cells (HSCs) in adult mammals. Prostate cancer commonly metastasizes to the bone and forms bone metastases in almost all patients who die of the disease. Prostate cancer bone metastases are thought to develop after rare bone marrow disseminated tumor cells (DTCs) escape a dormant state and reactivate. Prostate cancer DTCs and normal HSCs have been shown to compete for residence in the bone marrow and share many of same regulatory mechanisms for survival, proliferation and homing. In this review, we highlight these parallels in order to help our readers use the literature in HSC and DTC biology to inform their research and generate hypotheses in both fields.

Introduction

Prostate cancer (PCa) is a large public health problem with over 180,000 new cases and over 26,000 deaths per year in the United States alone [1]. Of PCa patients with distant metastases, 90% have metastases to bone [2]. PCa cells are thought to spread to the bone marrow early in the disease process, even at or before the time of curative intent surgery or radiation therapy to the prostate. At this time, they are termed disseminated tumor cells (DTCs) or micro-metastases. These cells are found in bone marrow or other tissues rather than circulating in peripheral blood, which most investigators term circulating tumor cells, or CTCs. The presence of bone marrow DTCs in PCa patients at the time of radical prostatectomy has been demonstrated by research groups at three institutions using various techniques including, RT-PCR for prostate specific antigen (PSA), immunohistochemistry for PSA and single cell selection of epithelial cell adhesion molecule (EPCAM) positive cells by single cell isolation after prior negative and positive selection [[3], [4], [5], [6], [7]]. Furthermore, their presence was shown by all three groups to correlate with PCa recurrence [5,8,9]. However, recently investigators reported being able to detect these cells in only a small minority of patients with localized PCa [10]. This report highlights the challenges in conducting this type of research and suggests further investigation.

PCa DTCs can remain viable but dormant for long time periods; as illustrated by the fact that about 20% of PCa recurrences after surgery occur greater than 5 years after patients were thought to be cured [11]. Reactivation or dormancy escape of bone marrow DTCs is thought to be a major cause of relapse in PCa and other malignancies [12]. Furthermore, an understanding of the biology of how DTCs or micrometastases are different from macroscopic tumors and how they interact with bone microenvironment has the potential to lend insight into later stages of the disease when metastases are visible on imaging. Therefore, understanding the biology of DTCs in the bone environment is crucial for prevention of relapse or treatment of relapsed disease.

There are two major models which have been invoked to describe tumor heterogeneity, either of which could describe the behavior of DTCs. The cancer stem cell (CSC) model suggests that subpopulations of cancer cells form a hierarchical cluster of tumor initiating cells [13]. These CSCs often feature parameters present in normal tissue stem cells, and like normal stem cells maintain themselves through self-renewal, but also differentiate into progenitor cell populations and ultimately into mature or non-stem cancer cells (NSCCs). The second major model for tumor development is the stochastic model. In the stochastic model, tumors develop by clonal evolution to progress into heterogeneous populations which are influenced by both intrinsic and extrinsic environmental factors [14].

Many tissues which harbor DTCs, including lung, liver and bone marrow, are also sites in which clinical relapse can eventually occur. Each of these organs support stem cell populations and tightly regulates proliferation as a component of normal function. This suggests that the normal activity of the host tissues to regulate stem cell function, be it the induction or maintenance of quiescence, ultimately becomes insufficient to enforce dormancy of DTCs. Yet, whether DTCs are primed for proliferation but constantly kept in check by suppressive signals or rather reprogrammed into a semi-permanent dormant state by the microenvironment remains unclear. In several experimental systems DTCs isolated directly from humans or from preclinical models are difficult to grow in vitro, and only proliferate after extended culture periods [12,15]. The implication from these studies is that DTCs, at least in these organs, are reprogrammed into a dormant state rather than held in check by the continual presence of negative regulators. Alternatively, there is ample evidence that immune regulation of DTCs plays a major role in controlling DTC proliferation [[16], [17], [18], [19]]. How the host is able to distinguish normal stem cells from tumor cells, be they CSCs or NSCCs, remains unclear.

We and others have hypothesized that DTCs reside in similar environments or “niches” as hematopoietic stem cells (HSCs) and compete for occupancy of the bone marrow microenvironment [20]. Likely because of shared interactions with stromal cell types, research over the past one or two decades has discovered shared regulatory mechanisms between DTCs and HSCs. In this review, we use HSC biology to frame a discussion of PCa bone marrow DTC regulation in hopes that we will help our readers gain intuition into DTC biology and help them generate new hypotheses by drawing on the more extensive HSC literature. We concentrate here on PCa, but note that many of the same concepts will apply to breast cancer and other malignancies which metastasize to bone. The mechanisms discussed below are summarized in Fig. 1.

Section snippets

Homing

Perhaps the most established and robust parallel between DTCs and HSCs is in homing to the bone marrow, much of which involves the cytokine CXCL12 (SDF-1). Over a decade ago, CXCL12 was suggested to be important for PCa bone metastasis [21]. Subsequently, blocking CXCL12 was shown to inhibit transit of PCa cells to the bone marrow. CXCL12 and was also shown to strongly co-localize with metastatic PCa cells – principally in the metaphysis of long bones in the mouse models used for these studies [

Location

We and others have shown that HSCs and DTCs functionally compete for residency in bone marrow [20]. In an analogous fashion to its function of maintaining HSCs in a pluripotent state, the bone marrow environment appears to cause PCa cells to assume a more primitive or cancer stem cell phenotype. Although this was long hypothesized, Shiozawa and colleagues recently showed a rapid assumption of a stem-like phenotype as defined by increased percentage of CD133+/CD44+ positive PCa cells [29].

Stress response and survival

A commonly proposed reason for “why” DTCs become dormant is as a survival mechanism [41]. After hematogenous spread to a new location such as the bone marrow, DTCs might not have the same proliferative signals present in the primary tumor and therefore stop cycling, which has been shown to correlate with resistance to chemotherapy and other causes of apoptosis. Prominent molecular mediators of this survival signaling include TGF-β2, p38 MAPK and the endoplasmic reticulum stress response [6,[41]

Dormancy vs. proliferation

Many of the best characterized regulators of PCa dormancy have analogous roles in regulation of HSC quiescence vs. proliferation. TGF-β2 is perhaps the best characterized factor maintaining dormancy in DTCs from PCa and other cancers [42]. Similarly, the TGFβ family member BMP-7 also maintains PCa DTC dormancy [46,47]. TGF-β family members have analogous effects for HSCs. TGF-β2 maintains HSCs in a quiescent state but increases their ability to engraft. Conversely, TGF-β1 inhibits HSC

Conclusions and perspective

Over the prior 20 years, we have seen remarkable parallels develop in our understanding of the biology of PCa DTCs and normal HSCs. This includes the study of localization, homing, survival, and proliferation. Frequently, the advances in normal hematology have preceded concurrent findings in solid tumor biology. This has provided enormous opportunities for hypothesis generation for investigators studying the biology of PCa DTCs. We are confident that these research parallels will continue in the

Financial support

Direct funding was provided by the NIH/NCI P01-CA093900, the NIH/NCI Tumor Microenvironment Network U54-CA163124 and supplement, Department of Defense W81XWH-14-1-0403 and W81XWH-15-1-0413. And Prostate Cancer Foundation Challenge award 16CHAL05. R.T. receives support as the Major McKinley Ash Colligate Professor. F.C. receives support from a Career Enhancement Award from the NIH/NCI Prostate Cancer Specialized Program in Research Excellence (SPORE) #F048931, sub-award of #F036250 at the

References (73)

  • S. Malladi et al.

    Metastatic latency and immune evasion through autocrine inhibition of WNT

    Cell

    (2016)
  • SEER Cancer Statistics Factsheets

    Prostate cancer. National Cancer Institute. Bethesda, MD

  • D.P. Wood et al.

    Identification of bone marrow micrometastases in patients with prostate cancer

    Cancer

    (1994)
  • S.W. Melchior et al.

    Early tumor cell dissemination in patients with clinically localized carcinoma of the prostate

    Clin. Cancer Res.

    (1997)
  • T.M. Morgan et al.

    Disseminated tumor cells in prostate cancer patients after radical prostatectomy and without evidence of disease predicts biochemical recurrence

    Clin. Cancer Res.

    (2009)
  • L. Chery et al.

    Characterization of single disseminated prostate cancer cells reveals tumor cell heterogeneity and identifies dormancy associated pathways

    Oncotarget

    (2014)
  • N.P. Murray et al.

    Differential expression of matrix metalloproteinase-2 expression in disseminated tumor cells and micrometastasis in bone marrow of patients with nonmetastatic and metastatic prostate cancer: theoretical considerations and clinical implications-an immunocytochemical study

    Bone Marrow Res.

    (2012)
  • D.P. Wood et al.

    Presence of circulating prostate cells in the bone marrow of patients undergoing radical prostatectomy is predictive of disease-free survival

    J. Clin. Oncol.

    (1997)
  • N.P. Murray et al.

    Minimum Residual disease in patients post radical prostatectomy for prostate cancer: theoretical considerations, clinical implications and treatment outcome

    Asian Pac. J. Cancer Prev.

    (2018)
  • H.J. Chalfin et al.

    Prostate cancer disseminated tumor cells are rarely detected in the bone marrow of localized patients undergoing radical prostatectomy across multiple rare cell detection platforms

    J. Urol.

    (2018)
  • C.R. Pound et al.

    Natural history of progression after PSA elevation following radical prostatectomy

    JAMA

    (1999)
  • M.S. Sosa et al.

    Mechanisms of disseminated cancer cell dormancy: an awakening field

    Nat. Rev. Cancer

    (2014)
  • M.S. Sosa et al.

    ERK1/2 and p38alpha/beta signaling in tumor cell quiescence: opportunities to control dormant residual disease

    Clin. Cancer Res.

    (2011)
  • B. Sheller et al.

    Analysis of a cancer dormancy model and control of immuno-therapy

    Math. Biosci. Eng.

    (2015)
  • H. Gonzalez et al.

    Innate and acquired immune surveillance in the post-dissemination phase of metastasis

    FEBS J.

    (2017)
  • L. Gelao et al.

    Tumour dormancy and clinical implications in breast cancer

    Ecancermedicalscience

    (2013)
  • C.N. Baxevanis et al.

    Cancer dormancy: a regulatory role for endogenous immunity in establishing and maintaining the tumor dormant state

    Vaccines (Basel)

    (2015)
  • Y. Shiozawa et al.

    Human prostate cancer metastases target the hematopoietic stem cell niche to establish footholds in mouse bone marrow

    J. Clin. Invest.

    (2011)
  • R.S. Taichman et al.

    Use of the stromal cell-derived factor-1/CXCR4 pathway in prostate cancer metastasis to bone

    Cancer Res.

    (2002)
  • Y.X. Sun et al.

    Skeletal localization and neutralization of the SDF-1(CXCL12)/CXCR4 axis blocks prostate cancer metastasis and growth in osseous sites in vivo

    J. Bone Miner. Res.

    (2005)
  • S. Mendez-Ferrer et al.

    Haematopoietic stem cell release is regulated by circadian oscillations

    Nature

    (2008)
  • Y. Jung et al.

    Annexin-2 is a regulator of stromal cell-derived factor-1/CXCL12 function in the hematopoietic stem cell endosteal niche

    Exp. Hematol.

    (2011)
  • Y. Jung et al.

    Annexin 2-CXCL12 interactions regulate metastatic cell targeting and growth in the bone marrow

    Mol. Cancer Res.

    (2015)
  • Y. Shiozawa et al.

    The marrow niche controls the cancer stem cell phenotype of disseminated prostate cancer

    Oncotarget

    (2016)
  • R.S. Taichman et al.

    Human osteoblasts support hematopoiesis through the production of granulocyte colony-stimulating factor

    J. Exp. Med.

    (1994)
  • L.M. Calvi et al.

    Osteoblastic cells regulate the haematopoietic stem cell niche

    Nature

    (2003)
  • Cited by (14)

    • Thorny ground, rocky soil: Tissue-specific mechanisms of tumor dormancy and relapse

      2022, Seminars in Cancer Biology
      Citation Excerpt :

      The HSC niche is powerfully quiescent, and HSCs can be found in endosteal [55–59], arteriolar [60], and sinusoidal [59,61,62] compartments under the influence of osteoblastic cells, MSCs, neurons, and endothelia. The HSC capacity for reversible quiescence while maintaining stemness is remarkably similar to properties of dormant DTCs, and therefore many studies have investigated parallel mechanisms of regulation (reviewed in [63]). Animal models show that the endosteal niche [19,40] and the perivascular niche [61,64–66] harbor myeloma, prostate, and breast cancer DTCs among others, and identifying the specific interactions that enable and regulate tumor dormancy is the next critical step.

    • Rebuilding the hematopoietic stem cell niche: Recent developments and future prospects

      2021, Acta Biomaterialia
      Citation Excerpt :

      The presence of dense vascular networks, different cells and a rich milieu of soluble factors makes the BM a preferable “soil” for tumor cells or the “seeds”. [250]. Several studies have drawn parallels between the homing of HSCs and tumor cell migration to the BM [251]. Signals from the primary tumor sites recruit supportive stromal components in the BM and lay the groundwork for tumor cell arrival, establishing the so-called “pre-metastatic niches”.

    • Disseminated tumor cells in bone marrow of cancer patients: Biology and clinical relevance

      2021, Bone Cancer: Bone Sarcomas and Bone Metastases - From Bench to Bedside
    • Abscisic acid regulates dormancy of prostate cancer disseminated tumor cells in the bone marrow

      2021, Neoplasia (United States)
      Citation Excerpt :

      These observations suggest that disseminated tumor cells (DTCs) may enter into a state of cellular dormancy for long periods within the bone marrow and possibly other sites [2–5]. Once in the marrow, DTC dormancy and subsequent reactivation are likely to be governed by systemic and local signals derived from the metastatic microenvironment [6,7]. Previously, we established that osteoblast-secreted growth arrest specific 6 (GAS6) signaling through TAM receptors (Tyro3, AXL, and MER) expressed on PCa cells regulates dormancy.

    View all citing articles on Scopus
    View full text