REVIEW

Bone metastasis of hepatocellular carcinoma: facts and hopes from clinical and translational perspectives

  • Zhao Huang 1,2,3 ,
  • Jingyuan Wen 1,2,3 ,
  • Yufei Wang 1,2,3 ,
  • Shenqi Han 1,2,3 ,
  • Zhen Li 4 ,
  • Xuemei Hu 4 ,
  • Dongling Zhu 5 ,
  • Zhenxiong Wang 6 ,
  • Junnan Liang 1,2,3 ,
  • Huifang Liang 1,2,3 ,
  • Xiao-ping Chen , 1,2,3,7 ,
  • Bixiang Zhang , 1,2,3,7
Expand
  • 1. Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
  • 2. Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan 430030, China
  • 3. Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
  • 4. Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
  • 5. Department of Nuclear Medicine and PET, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
  • 6. Department of Radiology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
  • 7. Key Laboratory of Organ Transplantation, Ministry of Education; Key Laboratory of Organ Transplantation, National Health Commission; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China

Received date: 25 Jan 2022

Accepted date: 21 Apr 2022

Published date: 15 Aug 2022

Copyright

2022 Higher Education Press

Abstract

Patients with hepatocellular carcinoma (HCC) and bone metastasis (BM) suffer from greatly reduced life quality and a dismal prognosis. However, BM in HCC has long been overlooked possibly due to its relatively low prevalence in previous decades. To date, no consensus or guidelines have been reached or formulated for the prevention and management of HCC BM. Our narrative review manifests the increasing incidence of HCC BM to sound the alarm for additional attention. The risk factors, diagnosis, prognosis, and therapeutic approaches of HCC BM are detailed to provide a panoramic view of this disease to clinicians and specialists. We further delineate an informative cancer bone metastatic cascade based on evidence from recent studies and point out the main factors responsible for the tumor-associated disruption of bone homeostasis and the formation of skeletal cancer lesions. We also present the advances in the pathological and molecular mechanisms of HCC BM to shed light on translational opportunities. Dilemmas and challenges in the treatment and investigation of HCC BM are outlined and discussed to encourage further endeavors in the exploration of underlying pathogenic and molecular mechanisms, as well as the development of novel effective therapies for HCC patients with BM.

Cite this article

Zhao Huang , Jingyuan Wen , Yufei Wang , Shenqi Han , Zhen Li , Xuemei Hu , Dongling Zhu , Zhenxiong Wang , Junnan Liang , Huifang Liang , Xiao-ping Chen , Bixiang Zhang . Bone metastasis of hepatocellular carcinoma: facts and hopes from clinical and translational perspectives[J]. Frontiers of Medicine, 2022 , 16(4) : 551 -573 . DOI: 10.1007/s11684-022-0928-z

1 Introduction

Bone is one of the most frequent metastatic destinations for cancers [1], and the mechanisms underlying the preferences for bone have attracted long-lasting interest that goes back for more than 100 years to Stephen Paget’s “seed and solid” hypothesis, which was proposed in 1889 [2]. Although skeleton involvement in breast, lung, and prostate cancers has received extensive attention, studies on the osseous metastasis of hepatocellular carcinoma (HCC) are quite limited.
The incidence of HCC bone metastasis (BM) continues to increase with the prolongation of overall survival time (OS) due to the development of novel diagnostic and therapeutic methods [3]. In fact, bone has become the second common metastatic site for HCC; BM accounts for approximately 30% of extrahepatic metastases and is second only to lung metastases [4]. In addition to the primary tumor burden, patients with BM suffer from skeleton-related events (SREs), loss of mobility, reduced quality of life, increased medical costs, and shortened OS [1]. The versatility and imperceptibility of HCC BM confuse diagnosis, and some patients with HCC are diagnosed with BM only at postmortem examination. The early diagnosis of HCC BM reduces the risk of fracture and paralysis, thus improving the quality of life of patients [5]. Imaging tools, including bone scintigraphy (BS), magnetic resonance imaging (MRI), computed tomography (CT), and positron emission tomography/computed tomography (PET/CT), provide access for the identification and surveillance of bone lesions [6]. The therapeutic options for HCC BM remain mainly confined to palliative treatments, such as external beam radiotherapy (EBRT), bone-targeting agents (BTAs), biologically targeted therapy, and surgery. Given that these strategies are rarely curative for HCC BM, disease progression prevention and palliative symptom reduction are the main goals [1]. In terms of advances in basic studies, the underlying mechanisms, as well as the molecular markers for the diagnosis and prognosis of HCC BM, have been investigated as discussed in the following sections. At the same time, the enriched and extended cancer bone metastatic cascade gleaned from other osteotropic cancers opens new avenues for profound studies on osteotropism in HCC. In addition, with the intensive investigation of immunomodulatory approaches for tumor treatment, the involvement of the bone immune-microenvironment is attracting increasing attention [7] and hints at the possibility of using immunomodulators, including anti-PD-1 agents [8], for curing HCC BM. Nevertheless, the translation of these findings into clinical applications still has a long way to go.
The importance of HCC BM remains underappreciated by clinicians, specialists, and the literature. The absence of a full overview of HCC bone metastatic events necessitates an up-to-date systematic narration. This review aims to illustrate the current status and future hopes for the treatment of HCC BM by comprehensively summarizing its clinical features and diagnostic and treatment approaches, as well as advances in basic studies on this disease.

2 Prevalence and risk factors of HCC BM

BM from HCC was uncommon and was previously described only in rare case reports or autopsies [9,10]. From 1962 to 2015, HCC BM varied from 1.97% to 23.59% in the incidence and accounted for 8.33%–37.09% of extrahepatic metastases (Tab.1). This discrepancy may be caused by the different diagnostic methods for HCC BM, diverse patient backgrounds, or statistical methods. Nevertheless, as evidenced by constitutive studies in the same centers [4,11] or the analyses of the same database [12,13], HCC BM has increased in prevalence over the past several decades (Tab.1). Less than one third of patients present with BM at the time of initial HCC diagnosis, and others develop BM during disease progression [14]. A total of 85% of patients with HCC and BM present with skeletal lesions as the primary extrahepatic disease manifestation [15].
Tab.1 Reported incidences of HCC BM in various patient cohorts
Period Center Diagnostic approaches Incidence of BM Rate of BM in EhMs
1962–1981 [16] Single Autopsy 16.10% (14/87) N.A.
1969–1978 [9] Single Autopsy 5.33% (12/225) 8.33% (12/144)
1969–1983 [4] Single Autopsy N.A. 20%
1978–1987 [11] Single BS, radiography, CT, and/or MRI 4.46% (12/269) N.A.
1983–1987 [10] Single Plain films, bone scans, or CT 5.06% (20/395) N.A.
1988–1997 [11] Single BS, radiography, CT, and/or MRI 12.87% (52/404) N.A.
1988–2012 [4] Single Autopsy N.A. 32.1%
1990–2005 [17] Single CT, MRI, BS, and/or PET with FDG 5.63% (56/995) 37.09% (56/151)
1990–2006 [18] Single Bone X-ray, BS, and CT or MRI 3.65% (87/2386) 25.44% (87/342)
1992–1997 [19] Single CT 10.17% (41/403) 27.70% (41/148)
2002–2014 [15] Single At least one imaging modality 23.59% (151/640) 32.90% (151/459)
2005–2015 [20] Single Symptoms, and CT or MRI 1.97% (20/1017) N.A.
2006–2009 [21] Single PET/CT N.A. 19.07% (49/257)
2009–2016 [5] Single Radiological images or pathological findings 9.71% (76/783) N.A.
2010–2013 [12] Multiple Unknown N.A. 22.93% (1008/4396)
2010–2014 [22] Multiple Unknown 4.29% (1567/36 507) N.A.
2010–2015 [13] Multiple Unknown N.A. 32.47% (1015/3126)

Note: Bolded information indicates constitutive analyses from the same institutions or database.Abbreviations: BM, bone metastasis; EhMs, extrahepatic metastases; N.A., not available; CT, computed tomography; MRI, magnetic resonance imaging; PET, positron emission tomography; FDG, fluorodeoxyglucose; BS, bone scintigraphy.

Multivariate logistic regression analyses have indicated that male sex, unmarried status, the high T/N stage of the primary tumor, the advanced histological grade of the primary tumor, high Child–Pugh score, lung metastases, intrahepatic metastases, and brain metastases are independent risk factors for BM diagnosis [14,15,22,23]. Additionally, HBV infection, insurance status, and high AFP levels are associated with the development of HCC BM [14,15].

3 Pathology and symptoms

HCC BM is mostly found in axial bones, such as vertebrae, the pelvis, and ribs [3,15,20]. The preferential skeletal localization sites of HCC metastases are found in areas of red marrow, which are rich in hematopoietic stem cells (HSCs), growth factors, and mineral substances and thus represent an active microenvironment for cellular growth [1]. Anatomically, concurrent portal hypertension and consequent collateral networks growing throughout the vertebral venous system in patients with HCC may be blamed for the premier propensity for BM to the axial skeleton [11,24]. More than half of the patients present with more than one skeletal lesion, and approximately 77.8% of BM cases are synchronous [14]. Histologically, osteolytic lesions of HCC BM account for 82.44% of all lesions, with 9.76% and 7.80% of lesions being osteoblastic or mixed, respectively [14]. Radiographically, HCC BM is destructive and expansive, with large hypervascular soft-tissue masses replacing the normal bone matrix [5,11].
In addition to the primary tumor burden, patients with BM mostly complain about bone pain [25], which is more likely caused by bone lesions (fractures, increased pressure on the endosteum, periosteum distortion, nerve root compression, and muscle spasms) rather than the tumor burden [26]. Patients also commonly develop SREs, such as pathological fractures, the need for radiotherapy (RT) to relieve bone pain or reduce structural damage within the bone, bone surgery to prevent or repair a fracture, spinal cord compression, and hypercalcemia, which often lead to impaired mobility, decreased quality of life, and an overwhelming medical burden [1]. More than half of the patients suffer from at least one SRE after the diagnosis of BM [14]. In patients with HCC, the RT of the affected bone is the leading SRE, followed by pathologic fracture, spinal cord compression, bone surgery, and hypercalcemia for the SREs throughout disease progression and for the first emerged SREs [14]. The percentages of patients with two and three SREs are 18.90% and 2.84%, respectively, whereby the median number of SREs is 2 [14]. The rates of occurrence of the first SRE within 1, 3, and 6 months after the diagnosis of BM are 30% (95% CI 22%–38%), 43% (95% CI 35%–52%), and 50% (95% CI 41%–59%), respectively. Patients who have an SRE are more susceptible to developing a second SRE after a median time of 0.9 months than those without [15]. The median times to the first, second, and third SREs after confirmed diagnosis of BM are 3, 6, and 9 months, respectively [14].

4 Imaging-based diagnostic approaches

The clinical symptoms and laboratory findings of patients with BM and HCC before the initiation of bone destruction or nerve depression are usually similar to those of patients with HCC but without BM [10,27]. Therefore, osseous involvement is frequently diagnosed when patients complain about pain in bone areas, bone fractures, or discomfort in the abdomen or undergo imaging examination during routine follow-up [27,28]. Imaging approaches are helpful for the early detection of BM and contribute to reduced rates of paralysis and bone fracture [6]. A meta-analysis conducted by Yang et al. concluded that different imaging techniques displayed distinct sensitivity and specificity [29]. For per-patient-based analysis, MRI (90.6%) and 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET) (89.7%) exhibited higher sensitivity than BS (86.0%) or CT (72.9%), whereas 18F-FDG PET (96.8%) together with CT (95.4%) and MRI (94.8%) were superior to BS (81.4%) in terms of specificity. For per-lesion based analysis, CT (77.1%), and BS (75.1%) showed inferior sensitivity than 18F-FDG PET (86.9%) or MRI (90.4%), whereas 18F-FDG PET (97.0%), MRI (96.0%), BS (93.6%), and CT (83.2%) showed decreased specificity.

4.1 Plain X-ray

Regardless of its relatively low sensitivity, plain X-ray is still recommended as one of the standard imaging methods for the diagnosis and evaluation of BM [1,6]. Rather than the cancer cells themselves, skeletal pathological changes are preconditions for the diagnosis of BM via X-ray given that they manifest as morphological changes in bone induced by cancer cells within the bone marrow (Fig.1). However, the sensitivity of X-ray for the BM of breast cancer in the skull, spine, and pelvis is limited to 44%–50% [30], and even metastases measuring up to 1 cm in the spongiosa of a vertebral body may be overlooked on plain X-ray [6]. Nevertheless, X-ray is of great importance, especially for detecting fractures and evaluating potential risks [6], because it is easily accessible and inexpensive.
Fig.1 Imaging techniques for the diagnosis of HCC BM. (A) X-ray images of metastatic HCC lesions in the right humerus (left image) and in the L1 vertebral body (right image). (B) Anterior and posterior positions of BS for a patient with multiple BM (left panel) and a patient with a bone lesion in the right scapula (right panel). (C) MRI of skeletal HCC lesions in the L1 vertebral body (upper row) and L3 vertebral body (lower row). (D) CT scans of BM in the right humerus (upper row) and the spine (lower row). (E) Whole-body 18F-FDG PET/CT images of BM in the fifth right rib (left panel) and right ilium (right panel). Arrows point to metastatic HCC bone lesions. Abbreviations: CT, computed tomography; BS, bone scintigraphy; MRI, magnetic resonance imaging; PET, positron emission tomography; BM, bone metastasis; LT, left; RT, right; AP, anteroposterior; TS, transverse section; T1WI, T1 weighted image; T2WI, T2 weighted image; STIR, short time inversion recovery; MIP, maximum intensity projection.

Full size|PPT slide

4.2 BS

BS, also referred to as bone scanning, is based on the accumulation of labeled phosphonates in bone areas with reactive hypermetabolism (Fig.1). This technique exhibits great sensitivity for osteosclerotic or mixed metastases, such as those in prostate cancer and breast cancer [6]. However, BS has inadequate sensitivity for dominantly osteolytic cancers, such as renal cell carcinoma, lymphoma, and HCC [11,31,32]. Furthermore, BS often fails to distinguish the flare phenomenon, which is caused by bone matrix regeneration and actually indicates successful treatment, from disease progression. Combination with single-photon emission computed tomography (SPECT) or SPECT/CT markedly increases the sensitivity and specificity of BS [33].

4.3 MRI

HCC BM commonly presents as hypervascular focal nodular masses with moderately intense enhancement on hepatic arterial dominant phase images and concomitantly moderately high signal intensity on fat-suppressed T1- and T2-weighted images. A study on HCC BM using the standard abdominal MRI protocol showed that BMs are best visualized at the arterial phase, and 56.4% show an arterial peak of enhancement; thus, early washout and arterial ring enhancement may be the specific signs of BM [34]. MRI can detect small BMs even when they are confined to the bone marrow (Fig.1) [35,36]. Most HCC BMs are located in the thoracic and lumbar spine, which are commonly included in the field-of-view of standard abdominal MRI protocols. This situation emphasizes that during routine follow-up scanning, radiologists should pay attention to the early diagnosis of BM in patients with HCC and high risks of bone involvement.

4.4 CT, PET, and PET/CT

CT can be used for the detection of osteolytic and osteoblastic lesions, as well as the assessment of the stabilities of bony structures (Fig.1) [6]. A CT scan has low sensitivity for tumors, except for very extensive tumors, restricted to the marrow space [6].
The combination of 18F-FDG PET with CT (PET/CT) can detect up to 80% of HCC skeletal metastases (Fig.1) [21,37] and is thus superior to BS, CT, and MRI [3841]. Additionally, 11Cacetate PET is more sensitive than 18F-FDG PET in patient-based and in lesion-based analyses [21,42]. The combined use of 11Cacetate and 18F-FDG as a dual tracer is due to the preference of 11Cacetate for well-differentiated tumors that complements the action of 18F-FDG in identifying the location of HCC [3]. In the evaluation of HCC metastasis, dual-tracer PET/CT displays higher sensitivity than single tracers [21,42]. The old bone tracer 18F sodium fluoride (18F-NaF) was recently rejuvenated with the help of PET. The accuracy of 18F-NaF PET/CT in the diagnosis of HCC BM is up to 95.7%, and the presence of 18F-NaF PET/CT-positive bone lesions predicts short survival in patients with HCC [43].

5 Prognosis and therapeutic approaches

The median OS time after the diagnosis of BM is dismal and ranges from 3.0 months to 11.7 months (Tab.2). The location, rather than the number, of bone lesions is the main factor associated with OS. Lesions in the spine are correlated with decreases in OS after the diagnosis of HCC and after the development of the first SRE [14,15]. Most patients die due to their intrahepatic HCC lesions instead of skeletal complications, indicating the importance of controlling intrahepatic tumors in patients with HCC whenever possible [ 11,17,44]. Aggressive treatments, including surgery for metastatic lesions, may produce a satisfactory local response and prolong OS [45]. Uka et al. recommended that most patients with HCC and extrahepatic metastases should mainly undergo treatment for the primary HCC tumor, although the treatment of extrahepatic metastases may improve survival in selected patients with HCC who have good hepatic reserve and low intrahepatic tumor stage (T0–T2) and are free of portal venous invasion [17]. To date, no guidelines or consensus has been established regarding the treatment strategy for HCC BM. Experiences from the guidelines for the management of BM in other cancers imply that treatment selection for patients with HCC and BM (Fig.2) should be based on the expected survival, symptoms, and condition of the patient, as well as the number and localization of bone lesions [46,47].
Tab.2 Prognosis and risk factors for HCC patients with BM
Period Treatment Prognosis   Independent risk factors
1978–1987 [11] RT, surgery, ethanol injection, and supportive care Median OS: 170 days   –
1981–1997 [48] Palliative RT Median OS from the diagnosis of spinal metastasis: 3 months   Responssive RT (complete   response, partial response)   and good performance   status (score < 2)
1988–1997 [11] RT, surgery, ethanol injection, and supportive care Median OS: 227 days   –
1991–2000 [25] RT OS rates at 1 and 2 years of 50% and 20%, respectively, with a median OS of 12 months from the time of HCC diagnosis; OS rates from the occurrence of BM at 1 and 2 years of 15% and 4%, respectively, with the median OS of 5 months   Tumor stage within the liver   and the presence of   metastases to organs
1992–2012 [49] RT Median OS from diagnosis of spinal metastases: 4.5 months; 1- and 2-year OS rates of 18.1% and 6.3%, respectively   Performance status (ECOG),   presence of uncontrolled   primary HCC, and presence   of extrahepatic metastases
1993–2013 [14] Sorafenib, RT, BP and surgery Median OS from the diagnosis of BM: 7 months   HCC etiology, performance   status (ECOG), BM   localized to the spine and   not receiving any BP   treatment
1997–2007 [44] RT Median OS after the diagnosis of BM: 7.4 months; 1-year and 2-year OS rates of 32.4% and 13.2%, respectively   Low KPS, high AFP levels,   uncontrolled intrahepatic   tumor, and receiving   treatment within the past 5   years
2000–2011 [50] RT Median OS: 7.0 months; OS rates at 1 and 2 years of 13.8% and 6.9%, respectively   –
2000–2018 [45] Surgery, RT, chemotherapy, and bone-modifying agents Median OS from the initiation of treatment: 7.4 ± 8.2 months (range 0.3–36 months) for all; 10.46 ± 8.05 months for surgical groups, and 5.19 ± 7.72 months for the conservative treatment groups   Patient’s general condition,   the serum albumin level,   and bone-modifying agent   treatment
2002–2009 [51] Irradiation/zoledronic acid Median OS from the initial date of therapy: 6.0 months (95% CI 0.0–12.7 months) for patients treated with zoledronic acid, and 4.2 months (95% CI 1.2–7.2 months) for patients treated with non-zoledronic acid; cumulative OS rates at 3 months of 74% and 44% and at 6 months of 79% and 37%   –
2002–2011 [52] SRS, cRT Median OS: 3 months in the cRT group and 7 months in the SRS group   Child–Pugh class and KPS
2002–2014 [15] Radiation, surgical resection, BPs, and sorafenib Median OS after the diagnosis of any type of metastasis: 5.6 months (95% CI 4.6–6.9)   AFP levels, Child–Pugh   score, and SREs
2005–2011 [53] EBRT Median OS after the first EBRT: 3.8 months   –
2006–2013 [54] Surgery, EBRT Median OS: 261 days (range 22–1359 days) after the diagnosis of metastasis, and 180 days (range 19–1351 days) after the initial operation   Tomita scoring system
2009–2014 [55] EBRT Median OS for the entire cohort: 8.0 months; 1-year and 2-year survival rates of 35.1% and 10.8%, respectively in patients receiving conventional fraction EBRT, and of 38.7% and 15.1%, respectively, in patients receiving hypofraction RT   KPS, TB, and intrahepatic   tumor control
2009–2016 [5] Sorafenib, sunitinib or lenvatinib RT, zoledronic acid and denosumab Median OS after the diagnosis of BM: 11.7 months (range 0.2–94.5 months)   Age over 75 years, HCV,   and Child–Pugh class B/C
2010–2014 [22] Surgery and other N/A Median OS from the time of diagnosis of HCC: 3.00 months (95% CI 2.77–3.24 months)   Unmarried, uninsured, high  primary tumor stage, high   regional lymph node (N1),   lung metastases, poor tumor   differentiated grade, and   elevated AFP, without surgery
2010–2014 [56] Zoledronic acid, palliative RT, curettage, and wide resection Median OS after BM diagnosis: 11 months (range 4–52); 1- and 2-year survival rates of 44.2% and 11.6%, respectively   Progression beyond the  University of California San   Francisco criteria and the   treatment of the primary   tumors
2011–2016 [57] RT Median OS after RT: 13.6, 4.8, and 2.6 months for the low-, intermediate-, and high-risk groups, respectively   ECOG performance status,   controlled primary HCC, and   extrahepatic metastases other   than bone
2014–2017 [58] RT and other N/A Median OS from the start of the RT for BM: 6.5 months; 1- and 2-year survival rates after diagnosis of BM of 35.5% and 13.5%, respectively   Child–Pugh class A group,   increase in AFP beyond 30 ng/mL,  and HCC size of   more than 5 cm
Abbreviations: RT, radiotherapy; OS, overall survival time; BM, bone metastases; ECOG, Eastern Cooperative Oncology Group; KPS, Karnofsky performance status; TB, total bilirubin; SRS, stereotactic radiosurgery; cRT, conventional radiation therapy; BP, bisphosphonate.
Fig.2 Outline of the multidisciplinary treatment options for patients with HCC and BM. Therapeutic strategies for HCC BM should be determined in accordance with the systematic evaluation of each patient’s general condition by a multidisciplinary team. BTAs are recommended to be started at the definite diagnosis of BM. Treatments for primary HCC, BM, and systematic symptoms are the three approaches for controlling disease progression and alleviating cancer-induced bone pain. Abbreviations: BM, bone metastases; BTAs, bone-targeting agents; EBRT, external beam radiotherapy; BPs, bisphosphonates; SRE, skeleton-related events.

Full size|PPT slide

5.1 Prognosis

Patients with HCC and BM have poor prognosis [13,15,22]. Survival estimates made by clinicians purely on the basis of experience are usually inaccurate [59]. Given the priority of expected OS in selecting therapeutic approaches to prevent overtreatment or undertreatment, numerous endeavors have been made to identify prognostic factors (Tab.2) or to develop prognostic models for patients with HCC and BM.
A prognostic nomogram encompassing RT, chemotherapy, and lung metastasis exhibited good accuracy for predicting OS, and its areas under the curve for 6-, 9-, and 12-month OS in the validation cohort were 0.698, 0.770, and 0.823, respectively [23]. A graded prognostic assessment for patients with HCC with spinal metastasis (HCC–SM GPA) consisting of Eastern Cooperative Oncology Group performance status, controlled primary HCC, and extrahepatic metastases other than bone, was used to stratify HCC–SM patients into low risk (GPA = 0), intermediate risk (GPA = 1 to 2), and high risk (GPA = 3 to 4) groups and successfully predicted survival outcomes in the validation cohort [57]. A prediction model for other cancer BM also provided available options. The Katagiri scoring system, which includes the primary lesion, visceral or cerebral metastases, abnormal laboratory data, poor performance status, previous chemotherapy, and multiple skeletal metastases, is suitable for OS estimation in patients with BM regardless of the location of skeletal involvement even after surgery and RT [60]. The models proposed by Bartels et al. [61], van der Linden et al. [62], or Bollen et al. [63], recommended by the Dutch national guidelines may also aid the choice of treatment.

5.2 RT

In analogy to BM in malignancies, such as breast and lung cancer, EBRT is frequently prescribed as palliative treatment for skeletal involvement in patients with HCC [55,64]. Although RT alone does not restore bone structure or stability, it is prescribed for relieving bone pain, preventing the worsening of the structural damage caused by BM, and reducing the need for subsequent surgery by controlling metastatic spread and preventing prosthesis displacement after surgery [1]. In patients with HCC and BM, EBRT results in an overall pain improvement in 73%–99.5% of cases and helps 17.0%–44.0% of patients achieve complete pain relief, thus reducing analgesic requirements [64].
Different radiation schedules have been proposed for common use in patients with BM [65]. Previous works showed that although no dose–response relationship existed for the overall response (OR), complete response (CR), and pain response rates of the palliative treatment of BM, the retreatment rate was higher in patients with expansile soft tissue receiving a lower dose of EBRT, whereas a higher radiological response rate and longer time to progression were achieved in high-dose RT groups [44,53,55,66]. Conventional RT led to a long duration of pain relief, whereas hypofractionated RT achieved early pain relief [55]. Similarly, a cohort study on 28 patients with HCC and BM reported that patients undergoing conventional high-dose multiple fractions (MFs) achieved pain relief for 5.0 months, which was significantly longer than the 1.8 and 3.0 months of pain relief experienced by those undergoing 8 Gy single fraction or moderate-dose MF therapy, respectively, but found no difference in the OR rates [50]. These results suggested that a high-dose MF schedule benefits patients with long predicted OS due to its long period of pain relief, whereas hypofractionated RT should be considered as an alternative for patients with short predicted survival times [55].
A prospective phase II study on 60 patients with gastrointestinal cancers (HCC, n = 25) and BM from 2014 to 2016 reported that RT in combination with zoledronic acid decreased the bone pain score from 6.7 to 2.8 at 1 month and to 2.1 at 3 months with the overall pain response rates of 95% and 96%. At the same time, the combination treatment improved the mean quality of life score from 66 to 56 and 55 at 1 and 3 months, respectively [67]. The addition of transcatheter arterial embolization (TAE) managed to shorten the mean time interval of pain relief from 15 days to 4.8 days and to reduce the recurrence rate of bone pain from 88% to 20% [68].

5.3 BTAs

BTAs mainly include bisphosphonates (BPs) and monoclonal antibodies. BPs are well known for reducing the frequency of SREs and bone pain and as a documented effective agent against malignant hypercalcemia in various cancers [1]. Three generations of BPs have been developed, with third-generation BPs, which include zoledronic acid, ibandronate, neridronate, and risedronate, exhibiting the strongest potency [69]. Zoledronic acid is currently the strongest nitrogen-containing BP and is used to treat cancer-induced osteolysis and thereby prevent skeletal complications associated with BM [70]. Zoledronic acid significantly delays the time to pain progression and shortens the time to radiographic progression in patients receiving non-RT for HCC with BM [51]. A case series consisting of 17 patients with BM from 2006 to 2008 reported that the visual analog scale decreased from 7.1 (±0.24) to 5.3 (±0.20) after the administration of zoledronic acid for at least three times; such an effect allowed a reduction in the use of analgesic drugs [71]. In addition to effects targeting osteoclasts, zoledronic acid suppresses HCC cell growth by inhibiting the translocation of Ras and RhoA, further terminating the mevalonate pathway and inducing the apoptosis of HCC cells [70].
Denosumab, an anti-RANKL antibody, is widely used to relieve bone pain and delay disease progression [1]. It has also been used for patients with HCC and BM [5,15,45]. In breast cancer and castration-resistant prostate cancer, denosumab has been proven to have superior efficacy over zoledronic acid as evidenced by the significant delay in the first and subsequent on-study SRE and the skeletal morbidity rate in the denosumab group relative to those in the zoledronic acid group [7274]. Moreover, Body et al. demonstrated that the suppression of bone resorption markers by denosumab is independent of prior BP treatment and of its biochemical efficacy. In patients with a poor biochemical response to BPs, denosumab displays superior performance with the significant suppression of bone resorption markers [75]. At the same time, denosumab was associated with low renal toxicity and few acute-phase reactions in a double-blind study for advanced breast cancer [72]. In terms of side effects, no significant difference in the rate of osteonecrosis of the jaw was found between denosumab and zoledronic acid [72,73].
Medical criteria for when BTA treatment should be started and stopped, especially for patients with HCC, are lacking. On the basis of experience with breast cancer, BTA treatment is recommended when skeletal involvement is diagnosed (Fig.2) and should be continued until the patient’s general performance status shows a substantial decline [76]. On the other hand, stopping or reducing the frequency of BTA treatment should be considered when the prognosis worsens and SRE-related biochemical markers of bone resorption decrease because this situation indicates that the osteotropic disease is not aggressive or is under control by the anticancer treatment [77].
To date, clinical trials on the BP zoledronic acid and the monoclonal antibody denosumab have excluded a large proportion of patients with advanced HCC [15]. Therefore, the benefits and side effects of these drugs in patients with HCC and BM still need further validation.

5.4 Sorafenib

In patients with advanced HCC, the multitarget receptor tyrosine kinase inhibitor sorafenib exhibits antitumor activity and is known to modestly improve survival over best supportive care [78]. Du et al. reported the case of a 74-year-old patient with HCC that had metastasized to the vertebrae and who responded to a reduced dose of sorafenib and subsequently demonstrated no signs of disease progression since starting treatment almost 5 years ago [79]. Sorafenib was also reported to protect against SREs [15,80].

5.5 Surgery

Although surgery for primary liver tumors significantly improves the survival of patients with BM, this result can potentially be explained by the biased selection of patients [22]. Surgery for bone metastatic lesions is controversial and is not predictive of prolonged survival according to some reports [13,23,81,82]. The median survival time of the patients with HCC and BM who underwent surgery is similar to that of patients who received only supportive care [11]. However, surgery relieves bone pain and reduces the incidence of SREs, thus improving the patients’ activities of daily living and life quality [45,81]. Even if a patient’s neurological function is compromised, surgery has the potential to reverse damage [81]. Surgery can maintain or recover ambulatory ability until recurrence or other spinal cord compression occurs, and a univariate analysis of the postoperative Frankel score has shown that patients who are ambulatory after surgery have better survival than those who become paraplegic [81].
The surgical indications and management specific to HCC BM are not different from those of other tumors [81]. The aim of surgery for BM is to stabilize pathological fractures, manage spinal cord compression, functionality and mobility, prevent impending fractures, and alleviate pain [1]. Patients with the life expectancy of at least 3 months are recommended to undergo minimally invasive procedures, whereas those with a life expectancy of 6 months can opt for open surgical procedures [46]. For spinal metastases, percutaneous techniques, such as vertebroplasty and kyphoplasty, are performed to stabilize the vertebrae, reduce microfracture-induced pain, and prevent the potential collapse of the vertebral body. These techniques do not reduce tumor size and should not be used in patients suffering from pain or neurological deficit caused by nerve root or spinal cord compression. Radiofrequency ablation is comparable with vertebroplasty, and surgeons often combine these two techniques to improve local relief [83,84]. Dorsal spinal decompression and stabilization are the standard surgical techniques for lumbar and thoracic metastases. The ventral/combined approach of decompression and ventral stable-angle plate osteosynthesis is commonly used for cervical lesions. A titanium mesh filled with bone cement can be applied to replace impaired vertebral bodies [81]. For long-bone metastases, intramedullary nailing with locking screws via minimally invasive techniques allows immediate full weight bearing, and a long-stem cemented endoprosthesis or a modular tumor endoprosthesis are alternatives for orthopedists [1]. Notably, preoperative low bone mineral density is an independent risk factor for cancer-specific mortality after hepatectomy for HCC [85].

5.6 TAE

Several studies have reported the beneficial effect of TAE in relieving bone pain or as a preoperative preparation for bone surgery [68,8688]. Uemura et al. showed that the mean time interval for initial pain relief was 4.7 days, which was shorter than the mean time interval in patients who received EBRT. At the same time, the combination of TAE and EBRT decreased the pain recurrence ratio from 75% to 20% [68].

5.7 Multidisciplinary teams and other palliative approaches

The specialists involved in a multidisciplinary team for the treatment of HCC BM should include the originally treating specialist, a radiation oncologist, a medical oncologist, and a radiologist. A neurologist should be present in the case of nerve root or spinal cord compression, and a neurosurgeon and/or orthopedic surgeon should participate in the case of possible operation indications [47].
Corticosteroids can be started in the case of symptomatic spinal cord compression to reduce swelling and edema around the spinal cord [46,47]. Analgesic drugs are usually unavoidable for pain management. The usage of analgesics, namely, non-narcotics, weak narcotics, and narcotics, should follow the three steps from the World Health Organization (WHO) guidelines all in association with adjuvant drugs [44,89].

6 Advances in basic studies

New findings on the osteotropism of various cancers precede the explorations of HCC BM and continue to provide the broad insights necessary for investigating the mystery and inspiring the studies on HCC BM. Thus, introducing the consensus and advances in cancer BM is critical.
To our knowledge, some prometastatic characteristics, such as the dynamic adhesive and migratory capabilities intrinsic to tumor cells, are common to the metastasis of most cancers, whereas some pathological steps and processes are specific to skeletal preference. For BM, the tumor-associated uncoupling of bone remodeling and the immunosuppressive microenvironment in the bone marrow are the essential prerequisites for the survival and thriving of cancer cells. These prerequisites will be our key points in the following discussion.

6.1 Cancer BM cascade

BM is a multistep process, and emerging findings have enriched our knowledge of the metastatic cascade. Osteotropism is a positive selection process at the primary site and in the bone, rather than passive adaption in the bone. Briefly, it encompasses premetastatic niche preparation, local invasion and intravasation, survival and spread in circulation, extravasation and adaption in the bone, micro- and macro-metastatic lesion formation, reciprocal communication with primary cancer, and further metastasis (Fig.3) [90,91].
Fig.3 Hypothetical bone metastatic cascade of HCC. On the basis of the academic advances in cancer BM, an integrated metastatic process was proposed for HCC bone lesion formation. (A) The malignant transformation of liver cells and microenvironment predispose HCC cells to metastasis by augmenting their mobility and inducing angiogenesis. HCC cells exploit the arterial bloodstream and vertebral venous system to spread to the bone cavity. The dysregulation and immunosuppressive status of the bone microenvironment benefit the settlement, proliferation, and further metastasis of disseminated tumor cells in the bone. Additionally, signals that induce the osteotropism of cancer cells are predelivered to the bone from the primary tumor site and prepare metastatic niches. Conversely, the bone microenvironment modulates the behavior of tumor cells at the primary site via the secretion of various bioactive substances. (B) The formation of a metastatic lesion in bone. The disseminated tumor cells first home to the bone cavity and adhere to bone stromal cells to settle inside the bone. Bone metastatic tumor cells either remain dormant or directly start to colonize and form micro-metastatic lesions. By interacting with various bone stromal cells, the migrated tumor cells finally thrive inside the bone and expand into a macrometastatic lesion.

Full size|PPT slide

Before tumor cells arrive at the bone, the bioactive substances secreted by primary cancer cells, such as cytokines and extracellular vesicles (EVs), instruct the bone marrow to prepare niches for tumor cell settlement [92]. The differentiated status and spatial localization of bone stromal cells in these niches provide advantages for the homing, adhesion, and colonization of the arriving metastatic cancer cells [9395]. For detachment from the primary site, primary cancer cells disrupt homeostasis by communicating with the surrounding stromal cells and degrading the extracellular matrix to achieve local invasion. Epithelial-to-mesenchymal transition (EMT) confers tumor cells with increased mobility, plasticity, self-renewal ability, and apoptosis resistance [96]. Invasive tumor cells enter the bloodstream for further spreading; this process is accompanied by angiogenesis. Circulating tumor cells (CTCs) suffer stress due to attack from the immune system and anoikis [97]. In fact, less than 1% of CTCs finally survive in the circulation and reach the distant metastatic destination [98101]. The size of the vasculature and the adhesive molecules of cancer cells largely determine their trapping in distant organs. For extravasation, given that the vasculature in the bone is highly fenestrated, bone metastatic tumor cells are more likely to penetrate the microvascular wall rather than rupture blood vessels by initiating intraluminal growth and forming an embolus [90]. The trapped CTCs then undergo mesenchymal-to-epithelial transition (MET) to settle and adhere to the bone marrow [96]. Bone-settled tumor cells either remain dormant or immediately interact with bone stromal cells to create a favorable environment for the formation of metastatic foci [90]. In addition to osteoclasts and osteoblasts, which are widely known for their involvement in a vicious cycle, other bone stromal cells, including osteocytes, bone marrow endothelial cells, hematopoietic cells, adipose stem cells, nerve cells, platelets, and myeloid and immune cells have been found to participate in cultivating “fertile soil” for metastatic “seeds” [94,95]. Importantly, although BM is the final step in bone metastatic cascades, the bone microenvironment can facilitate cancer cells in the bone to further metastasize and establish multiorgan secondary metastases [102]. Moreover, signaling from bone stromal cells to primary cancer cells can affect the progression of the primary tumor [85,103105]. In fact, the content of bone metastatic cascades continues to expand, and beyond the primary tumor and bone, other cells, organs, and systems may also be involved in cancer osteotropism. Cancer-associated fibroblasts (CAFs) in triple-negative breast cancer select bone metastatic seeds by skewing heterogeneous cancer cell populations toward a predominance of clones that thrive on the CAF-derived factors CXCL12 and IGF1, which are richly expressed in the bone microenvironment [106].

6.2 Uncoupled bone remodeling

Excessive bone remodeling processes lead to the release of growth factors and ionized calcium from the mineralized bone matrix. This effect promotes the growth of the metastatic tumor cells and the further secretion of osteolytic and osteoblastic factors, thus resulting in a vicious cycle. Tumor cells trigger this cycle by disrupting the balance between osteoclasts and osteoblasts through endocrine and paracrine factors (Fig.4).
Fig.4 Disturbed homeostasis of the bone microenvironment driven by tumor cells. Bone-metastasized tumor cells trigger the imbalances of immune-suppressive/immune-active and osteoclastic/osteoblastic activities to escape from immune elimination and initiate excessive bone reconstruction. Thriving tumor cells, the immune-privileged microenvironment, and uncoupled bone remodeling benefit each other through various bioactive molecules to perpetuate the destructive vicious cycle. Abbreviations: TAMs, tumor associated macrophages; Tregs, regulatory T cells; Th17, CD4+ T helper cells 17; MDSCs, myeloid-derived suppressor cells; DCs, dendritic cells; pDCs, plasmacytoid DCs; TI-DCs, tumor-infiltrating DCs; NK cells, natural killer cells; TIMs, tumor inflammatory macrophages.

Full size|PPT slide

Osteoclasts are polarized, multinucleated myeloid lineage cells that have differentiated from their mononuclear macrophage/monocyte lineage hematopoietic precursors. The activation of nuclear factor-κB, NFATc1, JUN N-terminal kinase, mitogen-activated protein kinase (MAPK), and phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathways promotes the formation and maturation of preosteoclasts [93,94]. Receptor activator for nuclear factor-κB ligand (RANKL) and macrophage colony stimulating factor are two important factors for the formation, activation, and function of osteoclasts that act by binding with their respective receptors, namely, RANK and colony stimulating factor 1 receptor, on preosteoclasts. Osteoprotegerin (OPG) is the endogenous decoy receptor that competes with RANK for RANKL and thereby inhibits osteoclastogenesis [93,94]. Some RANKL substitutes have been found to induce osteoclast formation in a RANKL-independent manner. Nontumor necrosis factor (TNF) superfamily growth factors, such as transforming growth factor β (TGFβ), interleukin-6, nerve growth factor (NGF), insulin-like growth factor (IGF) I, and IGF II induce osteoclast differentiation and activation. TNF superfamily growth factors, such as TNFα and TNF superfamily member 14; a proliferation inducing ligand (APRIL); and a B-cell activating factor (BAFF) belonging to the tumor necrosis factor family also generate significant numbers of tartrate-resistant acid-phosphatase-positive multinucleated cells, which can resorb bone. Notably, these noncanonical factors induce osteoclastogenesis in normal and pathological bone resorption [107]. PTHrP stimulates RANKL expression and inhibits OPG expression in osteoblasts, leading to malignant osteolysis [94]. Macrophage-stimulating protein binds to the RON receptor tyrosine kinase in osteoclasts, which is absent from osteoblasts, activating a RANK-independent, Src phosphorylation-dependent pathway to further stimulate osteoclast survival and activity [108].
Osteoblasts originate from mesenchymal stem cells (MSCs). Signals from bone morphogenetic proteins (BMPs), WNTs, TGFβ, endothelin 1 (ET1), insulin-like growth factors (IGFs), platelet-derived growth factor, urinary plasminogen activator, and fibroblast growth factors recruit the precursors of osteoblasts and accelerate their maturation [94]. During destructive bone remodeling, tumor cells suppress the formation and activity of osteoblasts by secreting activin A, the BMP inhibitor noggin, dickkopf-1, and sclerostin, thus aggravating the uncoupling between osteolytic and osteoblastic activities [95]. Transcriptional factors, including RUNX2, osterix, and activating transcription factor 4, drive the formation of osteoblasts and are used as markers for osteoblastogenesis.

6.3 Immunosuppressive bone microenvironment for tumor cells

During BM, tumor cells, local immune cells, and bone stromal cells intimately communicate with each other and perpetuate the vicious cycle (Fig.4) [109]. Although the bone is a major organ of the immune system, it appears to be an immunologically privileged environment for tumor cells for reasons that remain insufficiently understood. Several cell types and molecular signals, including the components of the innate and adaptive immune system, are responsible for the immunologically privileged status of cancer cells in bones. Natural killer (NK) cells, macrophages, regulatory T cells (Tregs), immature myeloid cells (IMCs), dendritic cells (DCs), and osteoblasts are well recognized pathological players and are of particular interest [7,110]. NK cells perform tumor-killing functions by inducing apoptosis through granule-mediated-exocytosis or Fas–Fas ligand interactions [111]. In bones, cancer cells express core2 β-1,6-N-acetylglucosaminyltransferase to escape NK cell-mediated apoptosis by disrupting the ligand-receptor-mediated (NKR/NKR-L and TRAIL/DR4) immune response [112]. Macrophages are mononuclear myeloid lineage cells that can be polarized into the M1-like subtype (proinflammatory tumor-suppressing macrophages) and the M2-like subtype (anti-inflammatory tumor-associated macrophages (TAMs)). Increased numbers of TAMs are found in prostate cancer with bone metastatic lesions, and macrophage depletion inhibits tumor growth inside the bone [113,114]. Moreover, chemokine (C–C motif) ligand 2 (CCL2)-expressing breast cancer cells recruit CCR2+ macrophages to prepare metastatic niches in lungs and bones [115]. These findings demonstrate that TAMs are closely associated with BM. Tregs are well known as CD4+ T cells that contribute to immune suppression, and bone marrow Tregs are significantly more abundant in bone metastatic prostate cancer than in other cancers [116]. In addition to anti-inflammatory cytokines, such as IL-10, IL-35, and TGFβ [110], RANK/RANKL signaling on DCs increases the number of Tregs [117119]. At the same time, CXCL12/C-X-C motif chemokine receptor (CXCR4) signaling promotes the trafficking of Tregs to the bone marrow [120]. CD4+ helper T cells (Th17 cells) are another important subset of CD4+ cells in cancer BM, and they also contribute to enhancing the activation of preosteoclasts and promoting osteolysis [121], Th17 cells can differentiate into Tregs under TGFβ stimulation during the immune response [122]. During the development of HSCs, a proportion of myeloid cells remains immature and executes an immunosuppressive function that is similar to the effect of Tregs [123]. IMCs, also referred to as myeloid-derived suppressor cells (MDSCs), are morphologically categorized into polymorphonuclear MDSCs (PMN-MDSCs) and monocytic MDSCs (M-MDSCs). PMN-MDSCs and M-MDSCs interfere with cytotoxic CD8+ T cells by generating reactive oxygen species and inducible NO synthase, and thereby promote bone metastatic seeding [124]. DC cells act as the key promoters of a functional cytotoxic T cell immune response via their antigen-presenting cell (APC) function. On the other hand, plasmacytoid DCs (pDCs) can recruit Tregs and MDSCs to promote tumor progression and metastasis [125], whereas tumor-infiltrating DCs can secrete TGFβ, nitric oxide, IL-10, VEGF, and arginase I to suppress the cytotoxic capacity of CD8+ cells [126]. The increased abundance of pDCs was found in the bone marrow of mice inoculated with breast cancer cells, and the deficiency of pDCs remarkably inhibited BM [127]. A group of osteoclasts prefers aerobic glycolysis, resulting in the accumulation of lactate. The lactate-rich environment benefits the growth of cancer cells, thus facilitating Treg function and suppressing the function of cytotoxic CD8+ T cells [128131]. Additionally, the expression of CXCL12 in osteoclasts recruits HSCs, Tregs, and metastatic seeds to the endosteum [116,120].

6.4 Molecular mechanisms of HCC BM

In addition to the common changes that contribute to BM, i.e., EMT, angiogenesis, and the vicious cycle inside the bone marrow [132], some intrinsic genetic alterations and molecular expression patterns that contribute to HCC BM (Fig.5) have been discovered (Tab.3) [133138].
Tab.3 Biomarkers for the prediction of diagnosis and prognosis of HCC BM
Gene Subject Expression pattern Biological function/molecular mechanism Potential application
CTGF [137,139] Primary tumor Highly expressed in patients with HCC and BM Risk factor of BM
IL-11 [137,139] Primary tumor Highly expressed in patients with HCC and BM Risk factor of BM
MMP-1 [137,140] Peritumor/HCC cell lines Highly expressed in patients with HCC and BM/in osteotropic cells lines Risk factor of BM
CXCR4 [139,141] Primary tumor Highly expressed in patients with HCC and BM Risk factor of BM
FRZB [136] BM Highly expressed in bone metastatic lesions
PNI [142] BM High density in bone metastatic lesions
RNF219 [134] Primary tumor/BM/HCC cell lines Highly expressed in patients with HCC and BM/in bone metastatic lesions/in osteotropic cell lines Promotes osteoclastogenesis by upregulating LGALS3 in a YAP1/β-catenin complex-dependent manner Therapeutic target
LGALS3 [134] Primary tumor/BM/HCC cell lines Highly expressed in patients with HCC and BM/in bone metastatic lesions/in osteotropic cell lines Promotes osteoclastogenesis and aggravate SREs Therapeutic target
miR-34a [143] Serum/primary tumor Low expression in the serum and in the primary tumor of patients with HCC and BM Promotes the migration and invasion of HCC cells by upregulating SMAD4 to further activate TGFβ signaling and upregulate its downstream effectors Risk factor of BM and therapeutic target
lnc34a [144] Serum/primary tumor Highly expressed in the serum and primary tumor of patients and HCC and BM Suppresses miR-34a expression epigenetically through DNMT3A/PHB2 and HDAC1 and sponging miR-34a Risk factor of BM
lncZEB1-AS1 [145] Primary tumor Highly expressed in patients with HCC and BM Promotes the migration and invasion of HCC cells by sponging miR-302b to activate PI3K/AKT signaling and increase EGFR expression Therapeutic target
H19 [133] Primary tumor/BM/HCC cell lines Highly expressed in patients with HCC and BM/in bone metastatic lesions/in osteotropic cell lines Promotes the migration and invasion of HCC cells by sponging miR-200b-3p and inducing osteoclastogenesis through the PPP1CA/p38MAPK axis Therapeutic target
CCL2 [146] CAFs Highly expressed in CAFs in primary site Promotes the migration of HCC cells by activating hedgehog signaling Therapeutic target
CCL5 [146] CAFs Highly expressed in CAFs in primary site Promotes the migration of HCC cells by activating hedgehog signaling Therapeutic target
CCL7 [146] CAFs Highly expressed in CAFs in primary site Promotes the invasion of HCC cells by activating TGFβ signaling Therapeutic target
CXCL16 [146] CAFs Highly expressed in CAFs in primary site Promotes the invasion of HCC cells by activating TGFβ signaling Therapeutic target
MAPK14 [147] BMECs Excessive activation in BMECs Upregulates ADAM17 expression Therapeutic target
ADAM17 [147] BMECs Highly expressed in BMECs Promotes the secretion of CX3CL1 Therapeutic target
CX3CL1 [147] HCC/BMECs Highly expressed in bone metastatic lesions/in BMECs Promotes the migration and invasion of HCC cells by activating PIK3CA/AKT1 and RHOA/ROCK2 signaling Therapeutic target
CX3CL1R [147] HCC Highly expressed in bone metastatic lesions Promotes the migration and invasion of HCC cells by activating PIK3CA/AKT1 and RHOA/ROCK2 signaling Therapeutic target

Abbreviations: HCC, hepatocellular carcinoma; BM, bone metastases; CTGF, connective tissue growth factor; IL-11, interleukin-11; MMP-1, matrix metallopeptidase-1; CXCR4, C-X-C motif chemokine receptor 4; FRZB, frizzled-related protein; PNI, perineural density; RNF219, ring finger protein 219; LGALS3, galectin 3; YAP1, Yes1 associated transcriptional regulator; SREs, skeleton-related events; SMAD4, SMAD family member 4; TGFβ, transforming growth factor β; DNMT3A, DNA methyltransferase 3 α; PHB2, prohibitin 2; HDAC1, histone deacetylase 1; lnc, long non-coding RNA; PI3K, phosphatidylinositol 3-kinase; AKT, AKT serine/threonine kinase; EGFR, epidermal growth factor receptor; PPP1CA, protein phosphatase 1 catalytic subunit α; MAPK, mitogen activated kinase-like protein; CCL, chemokine (C-C motif) ligand; CAFs, cancer-associated fibroblasts; CXCL, C-X-C motif chemokine ligand; BMECs, bone marrow endothelial cells; ADAM17, ADAM metallopeptidase domain 17; CX3CL1, C-X3-C motif chemokine ligand 1; CX3CL1R, CX3CL1 receptor; PIK3CA, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α; RHOA, ras homolog family member A; ROCK2, Rho associated coiled-coil containing protein kinase 2.

Fig.5 Molecular mechanism of HCC BM. Elevated expression levels of H19, lnc34a, lncZEB1, and RNF219 in HCC cells contribute to strengthened bone metastatic ability by enhancing the migration, invasion, and metastasis ability of HCC cells and inducing osteolytic activities in bone. CCL2, CCL5, CCL7, and CXCL16 secreted by CAFs in primary sites activate Hh and TGFβ signaling in HCC cells to facilitate their metastatic capacities. High levels of IL11, CTGF, and MMP-1 have been found in primary HCCs with bone lesions and predict bone metastatic events. Various cytokines released from bone remodeling and bone stromal cells, such as BMECs, support the outgrowth of disseminated HCC cells in bone. Abbreviations: CAFs, cancer-associated fibroblasts; BMEC, bone marrow endothelial cells; OC, osteoclasts.

Full size|PPT slide

Next-generation sequencing revealed mutations in two pairs of primary HCC and corresponding bone lesions. The deletions of ABL1, KIT, and EGFR or the missense mutations of CTNNB1, ERBB1, and KRAS were found in primary HCC and in paired BM, whereas the mutations of ALK, APC, and FGFR1 were observed only in primary HCC with mutations of DEAR, AKT1, and CDH1 being unique to HCC bone lesions [138]. A total of 22 genes in tumors and 45 genes in peritumor tissues were found to be differentially expressed between patients with HCC with and without BM. Among these genes, the high expressions of intratumoral connective tissue growth factor (CTGF), intratumoral interleukin-11 (IL-11), and peritumoral matrix metallopeptidase-1 (MMP-1) were independent prognostic factors for the progression to BM in patients with HCC [137]. CXCR4 was reported to be more highly expressed in patients with HCC and BM than in those without BM, and the high scores of CXCR4 in primary HCC predicted an increased risk for developing BM with decreased OS [141]. The same group that reported the above results further developed a clinicopathological prediction model consisting of vascular invasion, tumor–node–metastasis staging, CXCR4, CTGF, and IL-11, to predict the BM risk of patients with HCC [139]. Frizzled-related protein (FRZB) was found to be highly expressed in HCC specimens with BM and upregulated in bone metastatic lesions relative to in paired primary HCC tumor tissues, indicating that FRZB might play a key role in the BM of HCC [136]. Interestingly, they also revealed that perineural density was higher in HCC BM than in the corresponding primary liver tumor by using the same patient samples [142]. Patients with HCC and the high expression of RNF219 had shorter BM-free survival time than other patients. Mechanistically, the excessive activation of the RNF219/α-catenin/LGALS3 axis in HCC cells enhanced pro-osteolytic interactions with preosteoclasts, promoting osteoclastogenesis and BM formation, which in turn aggravated SREs. The administration of verteporfin, a small-molecule antagonist of the YAP–TEA interaction, decreased the RNF219-induced upregulation of LGALS3 and retarded RNF219/LGALS3-induced osteoclastogenesis, thus blocking the initiation and progression of HCC BM [134].
Noncoding RNAs also play a role in the HCC BM process. Higher serum levels of long noncoding RNA 34a (lnc34a), together with reduced serum and tumor levels of miR-34a, have been reported as independent risk factors for the development of BM in patients with BM [143,144]. lnc34a epigenetically decreases miR-34a expression by recruiting DNMT3A/PHB2 and HDAC1, which together sponge miR-34a [135]. The downregulation of miR-34a enhances the migration and invasion of cancer cells and activates the TGFβ-induced osteotropic transcription of the downstream genes by upregulating the expression of SMAD4 [135]. lncRNA zinc finger E-box binding homeobox 1 antisense 1 (lncZEB1-AS1) sponges miR-302b to activate PI3K/AKT signaling and increase EGFR expression, thereby promoting HCC BM. Univariate and multivariate analyses demonstrated that lncZEB1-AS1 is an independent risk factor for BM in patients with HCC [145]. In our own previous studies, we successfully established an animal model of HCC BM and isolated osteotropic HCC cell subpopulations by in vivo selection [133,140]. The expression levels of MMP-1, PTHrP, and CTGF were significantly elevated in osteotropic HCC cells relative to those in their ancestor cells. We further demonstrated that H19, a well-recognized lncRNA, promoted HCC BM by enhancing the mobility of HCC cells and modulating their interactions with osteoclasts. The high expression of H19 sponges miR-200b-3p to induce EMT, which confers HCC cells with increased mobility to spread from primary sites and reduces OPG expression by inactivating the p38MAPK signaling pathway, further enhancing the HCC-induced osteolytic process [133].
Beyond changes in liver cancer cells, alterations in the microenvironment at the primary site and bone marrow contribute to HCC progression and BM. CAFs in the liver promote HCC BM by secreting CCL2, CCL5, CCL7, and CXCL16 to activate hedgehog and TGFβ signaling [146]. The excessive activation of MAPK14 in bone marrow endothelial cells induces ADAM17-regulated CX3CL1 expression, which promotes the spinal metastasis of HCC cells with high CX3CLR expression by activating the PIK3CA/AKT1 and RHOA/ROCK2 signaling pathways [147].

7 Challenges and perspectives

Regardless of the increasing endeavors and impressive achievements in studies on HCC BM, the underlying pathological and molecular mechanisms, not to mention curative therapies, of this disease are far from clear. Here, we wish to emphasize the following four challenges facing the further investigation of HCC BM: ideal animal models, bone stromal cell involvement, novel drug targets, and effective drug delivery systems.
The limited access to clinical HCC BM specimens and ethical concerns emphasize the importance of an ideal and reproducible animal model for investigations on HCC BM. Given the extremely low incidence of spontaneous BM from orthotopic HCC, tumor cells are frequently directly injected into the bone cavity or into blood routes directed to bones. Specifically, the intraosseous access [148], left ventricle [140,148], vena cava [149], tail vein [150] and iliac artery [151,152] are commonly used for the introduction of cancer cells into the bone marrow. However, none of the models established through the above approaches can mimic all of the natural steps of HCC BM. Effective models should be able to reproduce the anatomical, genetic, and phenotypic changes during cancer cell diastasis from the primary site; survival in blood circulation; and settling and thriving in the bone marrow. To our knowledge, cancer cells in metastatic lesions acquire alterations that increase their tendency to metastasize to specific sites, and this preference can be strengthened by serial in vivo selection [153]. Thus, the preselection of HCC cell subpopulations with high propensity for BM may contribute to BM from orthotopic organs [154]. Specimens or organoids from patients with HCC and BM maintain the distinctive genomic alterations and functional heterogeneity of the original tissues that are prone to metastasize to the bone, and the xenografts of patient-derived specimens or organoids may contribute to spontaneous HCC BM in animal models [155,156].
The usage of immunodeficient mice for inoculating human cancer cells completely excludes immune cells, and the bone microenvironment in humans differs from that in mice. An animal model named NOD/SCID-hu, which is established by implanting human fetal bones or adult human ribs into nonobese diabetic/severe combined immunodeficient mice, realizes tissue- and species-specific metastasis [157]. In this animal model, the injected circulating prostate tumor cells form visible tumors only in human bone implants rather than in the mouse skeleton [158]. Humanized mice are also a promising tool for immune system investigation and for eliminating the discrepancy between humans and mice [159,160].
The bone marrow is a hemostatic organ with highly heterogeneous cells. Thus, beyond the interaction between HCC cells and osteoclasts or osteoblasts that have been intensively studied [133,134], other bone stromal cells deserve further attention. Studies have demonstrated that BMSCs can modulate HCC progression by transferring microRNAs through exosomes [105,161]. The involvement of the perineural system is implicated by the presence of more peripheral nerves in HCC bone lesions than in the corresponding primary liver tumor [142]. Studies on the BM of other tumor cells, bone marrow endothelial cells, hematopoietic cells, and adipose stem cells indicate that myeloid and immune cells may participate in HCC BM [94]. Additionally, combined single-cell and spatial transcriptomics have revealed the molecular, cellular, and spatial organization of the bone marrow niche; such a revelation opens a new avenue for the investigation of the communication between cancer cells and bone stromal cells [162]. Additional profound studies aimed at bone stromal cells are needed to obtain a further comprehensive understanding of HCC BM.
Another challenge is the efficient delivery of the therapeutic doses of drugs to tumor sites inside the bone marrow given the relatively low vascularization of bone tissue and the presence of physical barriers [151,163]. The administration of chemotherapeutic agents at high doses or frequency often leads to severe side effects and exerts dose-limiting toxicity on healthy tissues [164]. The affinity of BPs for the bone hydroxyapatite matrix has been utilized for the selective delivery of small-molecule drugs, cancer-targeting drugs and nanoparticles to the bone marrow [151,165]. Enclosing the BP-modified HER2 monoclonal antibody Tras or gold nanorods inside mesoporous silica nanoparticles results in the high efficiency of delivery to the bone marrow [151,166]. EVs have been established to mediate the communication among cells and have been revealed by various studies to be superior to conventional synthetic carriers in drug delivery [167]. Tumor-derived exosomes prepare pre-metastatic niches and determine organotropic metastasis via integrins [168]. Accordingly, exosomes or EVs with adequate organotropism are promising carriers for the delivery of bone- and tumor-targeted drugs to the bone marrow.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (Nos. 81372327, 81572427, and 81874189); the State Key Project on Infection Disease of China (No. 2018ZX10723204-003-003); the National Key Research and Development Program of China (No. 2018YFA0208904); the Major Technological Innovation Projects of Hubei Province (No. 2018ACA137); the Fundamental Research Funds for the Central Universities, HUST (No. 5001540059).

Compliance with ethics guidelines

Zhao Huang, Jingyuan Wen, Yufei Wang, Shenqi Han, Zhen Li, Xuemei Hu, Dongling Zhu, Zhenxiong Wang, Junnan Liang, Huifang Liang, Xiao-ping Chen, and Bixiang Zhang declare that they have no conflict of interest. This manuscript is a review article and does not involve a research protocol requiring approval by the relevant institutional review board or ethics committee.
1
Coleman RE, Croucher PI, Padhani AR, Clézardin P, Chow E, Fallon M, Guise T, Colangeli S, Capanna R, Costa L. Bone metastases. Nat Rev Dis Primers 2020; 6( 1): 83

DOI

2
Paget S. The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev 1989; 8( 2): 98– 101

3
Longo V, Brunetti O, D’Oronzo S, Ostuni C, Gatti P, Silvestris F. Bone metastases in hepatocellular carcinoma: an emerging issue. Cancer Metastasis Rev 2014; 33( 1): 333– 342

DOI

4
Schlageter M, Quagliata L, Matter M, Perrina V, Tornillo L, Terracciano L. Clinicopathological features and metastatic pattern of hepatocellular carcinoma: an autopsy study of 398 patients. Pathobiology 2016; 83( 6): 301– 307

DOI

5
Hirai T, Shinoda Y, Tateishi R, Asaoka Y, Uchino K, Wake T, Kobayashi H, Ikegami M, Sawada R, Haga N, Koike K, Tanaka S. Early detection of bone metastases of hepatocellular carcinoma reduces bone fracture and paralysis. Jpn J Clin Oncol 2019; 49( 6): 529– 536

DOI

6
Heindel W, Gübitz R, Vieth V, Weckesser M, Schober O, Schäfers M. The diagnostic imaging of bone metastases. Dtsch Arztebl Int 2014; 111( 44): 741– 747

7
Xiang L, Gilkes DM. The contribution of the immune system in bone metastasis pathogenesis. Int J Mol Sci 2019; 20( 4): 999

DOI

8
Wang K, Gu Y, Liao Y, Bang S, Donnelly CR, Chen O, Tao X, Mirando AJ, Hilton MJ, Ji RR. PD-1 blockade inhibits osteoclast formation and murine bone cancer pain. J Clin Invest 2020; 130( 7): 3603– 3620

DOI

9
Nakashima T, Okuda K, Kojiro M, Jimi A, Yamaguchi R, Sakamoto K, Ikari T. Pathology of hepatocellular carcinoma in Japan. 232 Consecutive cases autopsied in ten years. Cancer 1983; 51( 5): 863– 877

DOI

10
Liaw CC, Ng KT, Chen TJ, Liaw YF. Hepatocellular carcinoma presenting as bone metastasis. Cancer 1989; 64( 8): 1753– 1757

DOI

11
Fukutomi M, Yokota M, Chuman H, Harada H, Zaitsu Y, Funakoshi A, Wakasugi H, Iguchi H. Increased incidence of bone metastases in hepatocellular carcinoma. Eur J Gastroenterol Hepatol 2001; 13( 9): 1083– 1088

DOI

12
Oweira H, Petrausch U, Helbling D, Schmidt J, Mehrabi A, Schöb O, Giryes A, Abdel-Rahman O. Prognostic value of site-specific extra-hepatic disease in hepatocellular carcinoma: a SEER database analysis. Expert Rev Gastroenterol Hepatol 2017; 11( 7): 695– 701

DOI

13
Zhan H, Zhao X, Lu Z, Yao Y, Zhang X. Correlation and survival analysis of distant metastasis site and prognosis in patients with hepatocellular carcinoma. Front Oncol 2021; 11 : 652768

DOI

14
Santini D, Pantano F, Riccardi F, Di Costanzo GG, Addeo R, Guida FM, Ceruso MS, Barni S, Bertocchi P, Marinelli S, Marchetti P, Russo A, Scartozzi M, Faloppi L, Santoni M, Cascinu S, Maiello E, Silvestris F, Tucci M, Ibrahim T, Masi G, Gnoni A, Comandone A, Fazio N, Conti A, Imarisio I, Pisconti S, Giommoni E, Cinieri S, Catalano V, Palmieri VO, Infante G, Aieta M, Trogu A, Gadaleta CD, Brunetti AE, Lorusso V, Silvestris N. Natural history of malignant bone disease in hepatocellular carcinoma: final results of a multicenter bone metastasis survey. PLoS One 2014; 9( 8): e105268

DOI

15
Harding JJ, Abu-Zeinah G, Chou JF, Owen DH, Ly M, Lowery MA, Capanu M, Do R, Kemeny NE, O’Reilly EM, Saltz LB, Abou-Alfa GK. Frequency, morbidity, and mortality of bone metastases in advanced hepatocellular carcinoma. J Natl Compr Canc Netw 2018; 16( 1): 50– 58

DOI

16
Okazaki N, Yoshino M, Yoshida T, Hirohashi S, Kishi K, Shimosato Y. Bone metastasis in hepatocellular carcinoma. Cancer 1985; 55( 9): 1991– 1994

DOI

17
Uka K, Aikata H, Takaki S, Shirakawa H, Jeong SC, Yamashina K, Hiramatsu A, Kodama H, Takahashi S, Chayama K. Clinical features and prognosis of patients with extrahepatic metastases from hepatocellular carcinoma. World J Gastroenterol 2007; 13( 3): 414– 420

DOI

18
Uchino K, Tateishi R, Shiina S, Kanda M, Masuzaki R, Kondo Y, Goto T, Omata M, Yoshida H, Koike K. Hepatocellular carcinoma with extrahepatic metastasis: clinical features and prognostic factors. Cancer 2011; 117( 19): 4475– 4483

DOI

19
Katyal S, Oliver JH 3rd, Peterson MS, Ferris JV, Carr BS, Baron RL. Extrahepatic metastases of hepatocellular carcinoma. Radiology 2000; 216( 3): 698– 703

DOI

20
Bhatia R, Ravulapati S, Befeler A, Dombrowski J, Gadani S, Poddar N. Hepatocellular carcinoma with bone metastases: incidence, prognostic significance, and management-single-center experience. J Gastrointest Cancer 2017; 48( 4): 321– 325

DOI

21
Ho CL, Chen S, Cheng TK, Leung YL. PET/CT characteristics of isolated bone metastases in hepatocellular carcinoma. Radiology 2011; 258( 2): 515– 523

DOI

22
Guo X, Xu Y, Wang X, Lin F, Wu H, Duan J, Xiong Y, Han X, Baklaushev VP, Xiong S, Chekhonin VP, Peltzer K, Wang G, Zhang C. Advanced hepatocellular carcinoma with bone metastases: prevalence, associated factors, and survival estimation. Med Sci Monit 2019; 25 : 1105– 1112

DOI

23
Hu C, Yang J, Huang Z, Liu C, Lin Y, Tong Y, Fan Z, Chen B, Wang C, Zhao CL. Diagnostic and prognostic nomograms for bone metastasis in hepatocellular carcinoma. BMC Cancer 2020; 20( 1): 494

DOI

24
Coleman RE. Clinical features of metastatic bone disease and risk of skeletal morbidity. Clin Cancer Res 2006; 12( 20): 6243s– 6249s

DOI

25
Seong J, Koom WS, Park HC. Radiotherapy for painful bone metastases from hepatocellular carcinoma. Liver Int 2005; 25( 2): 261– 265

DOI

26
Goblirsch MJ, Zwolak PP, Clohisy DR. Biology of bone cancer pain. Clin Cancer Res 2006; 12( 20): 6231s– 6235s

DOI

27
Monteserin L, Mesa A, Fernandez-Garcia MS, Gadanon-Garcia A, Rodriguez M, Varela M. Bone metastases as initial presentation of hepatocellular carcinoma. World J Hepatol 2017; 9( 29): 1158– 1165

DOI

28
Doval DC, Bhatia K, Vaid AK, Pavithran K, Sharma JB, Hazarika D, Jena A. Spinal cord compression secondary to bone metastases from hepatocellular carcinoma. World J Gastroenterol 2006; 12( 32): 5247– 5252

29
Yang HL, Liu T, Wang XM, Xu Y, Deng SM. Diagnosis of bone metastases: a meta-analysis comparing 18FDG PET, CT, MRI and BS. Eur Radiol 2011; 21( 12): 2604– 2617

DOI

30
Hamaoka T, Madewell JE, Podoloff DA, Hortobagyi GN, Ueno NT. Bone imaging in metastatic breast cancer. J Clin Oncol 2004; 22( 14): 2942– 2953

DOI

31
Chen CY, Wu K, Lin WH, Lan TY, Wang SY, Sun JS, Weng PW, Yen RF, Yang RS. High false negative rate of Tc-99m MDP whole-body BS in detecting skeletal metastases for patients with hepatoma. J Formos Med Assoc 2012; 111( 3): 140– 146

DOI

32
Jin YJ, Lee HC, Lee D, Shim JH, Kim KM, Lim YS, Do KH, Ryu JS. Role of the routine use of chest computed tomography and bone scan in staging workup of hepatocellular carcinoma. J Hepatol 2012; 56( 6): 1324– 1329

DOI

33
Chua S, Gnanasegaran G, Cook GJ. Miscellaneous cancers (lung, thyroid, renal cancer, myeloma, and neuroendocrine tumors): role of SPECT and PET in imaging bone metastases. Semin Nucl Med 2009; 39( 6): 416– 430

DOI

34
Velloni F, Ramalho M, AlObaidy M, Matos AP, Altun E, Semelka RC. Bone metastases of hepatocellular carcinoma: appearance on MRI using a standard abdominal protocol. AJR Am J Roentgenol 2016; 206( 5): 1003– 1012

DOI

35
Asenbaum U, Nolz R, Karanikas G, Furtner J, Woitek R, Simonitsch-Klupp I, Raderer M, Mayerhoefer ME. Bone marrow involvement in malignant lymphoma: evaluation of quantitative PET and MRI BIomarkers. Acad Radiol 2018; 25( 4): 453– 460

DOI

36
Rashidi A Baratto L Theruvath AJ Greene EB Hawk KE Lu R Link MP Spunt SL Daldrup-Link HE. Diagnostic accuracy of 2-[18F]FDG-PET and whole-body DW-MRI for the detection of bone marrow metastases in children and young adults . Eur Radiol 2022; [Epub ahead of print] doi:10.1007/s00330-021-08529-x

37
Sugiyama M, Sakahara H, Torizuka T, Kanno T, Nakamura F, Futatsubashi M, Nakamura S. 18F-FDG PET in the detection of extrahepatic metastases from hepatocellular carcinoma. J Gastroenterol 2004; 39( 10): 961– 968

DOI

38
Yoon KT Kim JK Kim DY Ahn SH Lee JD Yun M Rha SY Chon CY Han KH. Role of 18F-fluorodeoxyglucose positron emission tomography in detecting extrahepatic metastasis in pretreatment staging of hepatocellular carcinoma . Oncology 2007; 72(Suppl 1): 104– 110

39
Kim YK, Lee KW, Cho SY, Han SS, Kim SH, Kim SK, Park SJ. Usefulness 18F-FDG positron emission tomography/computed tomography for detecting recurrence of hepatocellular carcinoma in posttransplant patients. Liver Transpl 2010; 16( 6): 767– 772

DOI

40
Fujimoto R, Higashi T, Nakamoto Y, Hara T, Lyshchik A, Ishizu K, Kawashima H, Kawase S, Fujita T, Saga T, Togashi K. Diagnostic accuracy of bone metastases detection in cancer patients: comparison between bone scintigraphy and whole-body FDG-PET. Ann Nucl Med 2006; 20( 6): 399– 408

DOI

41
Nagaoka S, Itano S, Ishibashi M, Torimura T, Baba K, Akiyoshi J, Kurogi J, Matsugaki S, Inoue K, Tajiri N, Takada A, Ando E, Kuromatsu R, Kaida H, Kurogi M, Koga H, Kumashiro R, Hayabuchi N, Kojiro M, Sata M. Value of fusing PET plus CT images in hepatocellular carcinoma and combined hepatocellular and cholangiocarcinoma patients with extrahepatic metastases: preliminary findings. Liver Int 2006; 26( 7): 781– 788

DOI

42
Ho CL, Chen S, Yeung DW, Cheng TK. Dual-tracer PET/CT imaging in evaluation of metastatic hepatocellular carcinoma. J Nucl Med 2007; 48( 6): 902– 909

DOI

43
Yen RF, Chen CY, Cheng MF, Wu YW, Shiau YC, Wu K, Hong RL, Yu CJ, Wang KL, Yang RS. The diagnostic and prognostic effectiveness of F-18 sodium fluoride PET-CT in detecting bone metastases for hepatocellular carcinoma patients. Nucl Med Commun 2010; 31( 7): 637– 645

DOI

44
He J, Zeng ZC, Tang ZY, Fan J, Zhou J, Zeng MS, Wang JH, Sun J, Chen B, Yang P, Pan BS. Clinical features and prognostic factors in patients with bone metastases from hepatocellular carcinoma receiving external beam radiotherapy. Cancer 2009; 115( 12): 2710– 2720

DOI

45
Uei H, Tokuhashi Y, Maseda M. Treatment outcomes of patients with spinal metastases derived from hepatocellular carcinoma. Int J Clin Oncol 2018; 23( 5): 886– 893

DOI

46
Bollen L, Dijkstra SPD, Bartels RHMA, de Graeff A, Poelma DLH, Brouwer T, Algra PR, Kuijlen JMA, Minnema MC, Nijboer C, Rolf C, Sluis T, Terheggen MAMB, van der Togt-van Leeuwen ACM, van der Linden YM, Taal W. Clinical management of spinal metastases—The Dutch national guideline. Eur J Cancer 2018; 104 : 81– 90

DOI

47
Sodji Q, Kaminski J, Willey C, Kim N, Mourad W, Vender J, Dasher B. Management of metastatic spinal cord compression. South Med J 2017; 110( 9): 586– 593

DOI

48
Chang SS, Luo JC, Chao Y, Chao JY, Chi KH, Wang SS, Chang FY, Lee SD, Yen SH. The clinical features and prognostic factors of hepatocellular carcinoma patients with spinal metastasis. Eur J Gastroenterol Hepatol 2001; 13( 11): 1341– 1345

DOI

49
Choi C, Seong J. Predictive factors of palliative radiotherapy response and survival in patients with spinal metastases from hepatocellular carcinoma. Gut Liver 2015; 9( 1): 94– 102

DOI

50
Hayashi S, Tanaka H, Hoshi H. External beam radiotherapy for painful bone metastases from hepatocellular carcinoma: multiple fractions compared with an 8-Gy single fraction. Nagoya J Med Sci 2014; 76( 1-2): 91– 99

51
Katamura Y, Aikata H, Hashimoto Y, Kimura Y, Kawaoka T, Takaki S, Waki K, Hiramatsu A, Kawakami Y, Takahashi S, Kenjo M, Chayama K. Zoledronic acid delays disease progression of bone metastases from hepatocellular carcinoma. Hepatol Res 2010; 40( 12): 1195– 1203

DOI

52
Chang UK, Kim MS, Han CJ, Lee DH. Clinical result of stereotactic radiosurgery for spinal metastasis from hepatocellular carcinoma: comparison with conventional radiation therapy. J Neurooncol 2014; 119( 1): 141– 148

DOI

53
Kim T, Cha HJ, Kim JW, Seong J, Lee IJ. High dose and compartmental target volume may improve patient outcome after radiotherapy for pelvic bone metastases from hepatocellular carcinoma. Oncotarget 2016; 7( 33): 53921– 53929

DOI

54
Lee MH, Lee SH, Kim ES, Eoh W, Chung SS, Lee CS. Survival-related factors of spinal metastasis with hepatocellular carcinoma in current surgical treatment modalities: a single institute experience. J Korean Neurosurg Soc 2015; 58( 5): 448– 453

DOI

55
He J, Shi S, Ye L, Ma G, Pan X, Huang Y, Zeng Z. A randomized trial of conventional fraction versus hypofraction radiotherapy for bone metastases from hepatocellular carcinoma. J Cancer 2019; 10( 17): 4031– 4037

DOI

56
Lu Y, Hu JG, Lin XJ, Li XG. Bone metastases from hepatocellular carcinoma: clinical features and prognostic factors. Hepatobiliary Pancreat Dis Int 2017; 16( 5): 499– 505

DOI

57
Rim CH, Choi C, Choi J, Seong J. Establishment of a disease-specific graded prognostic assessment for hepatocellular carcinoma patients with spinal metastasis. Gut Liver 2017; 11( 4): 535– 542

DOI

58
Kim S, Choi Y, Kwak DW, Lee HS, Hur WJ, Baek YH, Lee SW. Prognostic factors in hepatocellular carcinoma patients with bone metastases. Radiat Oncol J 2019; 37( 3): 207– 214

DOI

59
Gwilliam B, Keeley V, Todd C, Roberts C, Gittins M, Kelly L, Barclay S, Stone P. Prognosticating in patients with advanced cancer—observational study comparing the accuracy of clinicians’ and patients’ estimates of survival. Ann Oncol 2013; 24( 2): 482– 488

DOI

60
Kubota H, Soejima T, Sulaiman NS, Sekii S, Matsumoto Y, Ota Y, Tsujino K, Fujita I, Fujimoto T, Morishita M, Ikegaki J, Matsumoto K, Sasaki R. Predicting the survival of patients with bone metastases treated with radiation therapy: a validation study of the Katagiri scoring system. Radiat Oncol 2019; 14( 1): 13

DOI

61
Bartels RH, Feuth T, van der Maazen R, Verbeek AL, Kappelle AC, André Grotenhuis J, Leer JW. Development of a model with which to predict the life expectancy of patients with spinal epidural metastasis. Cancer 2007; 110( 9): 2042– 2049

DOI

62
van der Linden YM, Dijkstra SP, Vonk EJ, Marijnen CA, Leer JW; Dutch Bone Metastasis Study Group. Prediction of survival in patients with metastases in the spinal column: results based on a randomized trial of radiotherapy. Cancer 2005; 103( 2): 320– 328

DOI

63
Bollen L, van der Linden YM, Pondaag W, Fiocco M, Pattynama BP, Marijnen CA, Nelissen RG, Peul WC, Dijkstra PD. Prognostic factors associated with survival in patients with symptomatic spinal bone metastases: a retrospective cohort study of 1,043 patients. Neuro-oncol 2014; 16( 7): 991– 998

DOI

64
Hayashi S, Tanaka H, Hoshi H. Palliative external-beam radiotherapy for bone metastases from hepatocellular carcinoma. World J Hepatol 2014; 6( 12): 923– 929

DOI

65
Rutter CE, Yu JB, Wilson LD, Park HS. Assessment of national practice for palliative radiation therapy for bone metastases suggests marked underutilization of single-fraction regimens in the United States. Int J Radiat Oncol Biol Phys 2015; 91( 3): 548– 555

DOI

66
Jung IH, Yoon SM, Kwak J, Park JH, Song SY, Lee SW, Ahn SD, Choi EK, Kim JH. High-dose radiotherapy is associated with better local control of bone metastasis from hepatocellular carcinoma. Oncotarget 2017; 8( 9): 15182– 15192

DOI

67
Choi J, Lee EJ, Yang SH, Im YR, Seong J. A prospective phase II study for the efficacy of radiotherapy in combination with zoledronic acid in treating painful bone metastases from gastrointestinal cancers. J Radiat Res (Tokyo) 2019; 60( 2): 242– 248

DOI

68
Uemura A, Fujimoto H, Yasuda S, Osaka I, Goto N, Shinozaki M, Ito H. Transcatheter arterial embolization for bone metastases from hepatocellular carcinoma. Eur Radiol 2001; 11( 8): 1457– 1462

DOI

69
Barbosa JS, Almeida Paz FA, Braga SS. Bisphosphonates, old friends of bones and new trends in clinics. J Med Chem 2021; 64( 3): 1260– 1282

DOI

70
Honda Y, Takahashi S, Zhang Y, Ono A, Murakami E, Shi N, Kawaoka T, Miki D, Tsuge M, Hiraga N, Abe H, Ochi H, Imamura M, Aikata H, Chayama K. Effects of bisphosphonate zoledronic acid in hepatocellular carcinoma, depending on mevalonate pathway. J Gastroenterol Hepatol 2015; 30( 3): 619– 627

DOI

71
Montella L, Addeo R, Palmieri G, Caraglia M, Cennamo G, Vincenzi B, Guarrasi R, Mamone R, Faiola V, Frega N, Capasso E, Maiorino L, Leopardo D, Pizza C, Montesarchio V, Del Prete S. Zoledronic acid in the treatment of bone metastases by hepatocellular carcinoma: a case series. Cancer Chemother Pharmacol 2010; 65( 6): 1137– 1143

DOI

72
Stopeck AT, Lipton A, Body JJ, Steger GG, Tonkin K, de Boer RH, Lichinitser M, Fujiwara Y, Yardley DA, Viniegra M, Fan M, Jiang Q, Dansey R, Jun S, Braun A. Denosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast cancer: a randomized, double-blind study. J Clin Oncol 2010; 28( 35): 5132– 5139

DOI

73
Fizazi K, Carducci M, Smith M, Damião R, Brown J, Karsh L, Milecki P, Shore N, Rader M, Wang H, Jiang Q, Tadros S, Dansey R, Goessl C. Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study. Lancet 2011; 377( 9768): 813– 822

DOI

74
Lipton A, Siena S, Rader M, Bilynskyy B, Viniegra M, Richardson G, Beuzeboc P, Clemens M, Ke C, Jun S. Comparison of denosumab versus zoledronic acid (Za) for treatment of bone metastases in advanced cancer patients: an integrated analysis of 3 pivotal trials. Ann Oncol 2010; 21 : 380

75
Body JJ, Lipton A, Gralow J, Steger GG, Gao G, Yeh H, Fizazi K. Effects of denosumab in patients with bone metastases with and without previous bisphosphonate exposure. J Bone Miner Res 2010; 25( 3): 440– 446

DOI

76
Hillner BE, Ingle JN, Chlebowski RT, Gralow J, Yee GC, Janjan NA, Cauley JA, Blumenstein BA, Albain KS, Lipton A, Brown S; American Society of Clinical Oncology. American Society of Clinical Oncology 2003 update on the role of bisphosphonates and bone health issues in women with breast cancer. J Clin Oncol 2003; 21( 21): 4042– 4057

DOI

77
Coleman RE, Major P, Lipton A, Brown JE, Lee KA, Smith M, Saad F, Zheng M, Hei YJ, Seaman J, Cook R. Predictive value of bone resorption and formation markers in cancer patients with bone metastases receiving the bisphosphonate zoledronic acid. J Clin Oncol 2005; 23( 22): 4925– 4935

DOI

78
Su GL, Altayar O, O’Shea R, Shah R, Estfan B, Wenzell C, Sultan S, Falck-Ytter Y. AGA Clinical Practice Guideline on Systemic Therapy for Hepatocellular Carcinoma. Gastroenterology 2022; 162( 3): 920– 934

DOI

79
Du J, Qian X, Liu B. Long-term progression-free survival in a case of hepatocellular carcinoma with vertebral metastasis treated with a reduced dose of sorafenib: case report and review of the literature. Oncol Lett 2013; 5( 1): 381– 385

DOI

80
Harding JJ, Abou-Alfa GK. Treating advanced hepatocellular carcinoma: how to get out of first gear. Cancer 2014; 120( 20): 3122– 3130

DOI

81
Zhang D, Xu W, Liu T, Yin H, Yang X, Wu Z, Xiao J. Surgery and prognostic factors of patients with epidural spinal cord compression caused by hepatocellular carcinoma metastases: retrospective study of 36 patients in a single center. Spine 2013; 38( 17): E1090– E1095

DOI

82
Cho HS, Oh JH, Han I, Kim HS. Survival of patients with skeletal metastases from hepatocellular carcinoma after surgical management. J Bone Joint Surg Br 2009; 91( 11): 1505– 1512

DOI

83
van der Linden E, Kroft LJ, Dijkstra PD. Treatment of vertebral tumor with posterior wall defect using image-guided radiofrequency ablation combined with vertebroplasty: preliminary results in 12 patients. J Vasc Interv Radiol 2007; 18( 6): 741– 747

DOI

84
Zheng L, Chen Z, Sun M, Zeng H, Zuo D, Hua Y, Cai Z. A preliminary study of the safety and efficacy of radiofrequency ablation with percutaneous kyphoplasty for thoracolumbar vertebral metastatic tumor treatment. Med Sci Monit 2014; 20 : 556– 563

DOI

85
Miyachi Y, Kaido T, Yao S, Shirai H, Kobayashi A, Hamaguchi Y, Kamo N, Yagi S, Uemoto S. Bone mineral density as a risk factor for patients undergoing surgery for hepatocellular carcinoma. World J Surg 2019; 43( 3): 920– 928

DOI

86
Facchini G, Di Tullio P, Battaglia M, Bartalena T, Tetta C, Errani C, Mavrogenis AF, Rossi G. Palliative embolization for metastases of the spine. Eur J Orthop Surg Traumatol 2016; 26( 3): 247– 252

DOI

87
Kim W, Han I, Jae HJ, Kang S, Lee SA, Kim JS, Kim HS. Preoperative embolization for bone metastasis from hepatocellular carcinoma. Orthopedics 2015; 38( 2): e99– e105

DOI

88
Ma J, Tullius T, Van Ha TG. Update on preoperative embolization of bone metastases. Semin Intervent Radiol 2019; 36( 3): 241– 248

DOI

89
Sakaguchi M, Maebayashi T, Aizawa T, Ishibashi N, Fukushima S, Saito T. Radiation therapy and palliative care prolongs the survival of hepatocellular carcinoma patients with bone metastases. Intern Med 2016; 55( 9): 1077– 1083

DOI

90
Obenauf AC, Massagué J. Surviving at a distance: organ-specific metastasis. Trends Cancer 2015; 1( 1): 76– 91

DOI

91
Satcher RL, Zhang XH. Evolving cancer-niche interactions and therapeutic targets during bone metastasis. Nat Rev Cancer 2022; 22( 2): 85– 101

DOI

92
Wang H, Zhang W, Bado I, Zhang XH. Bone tropism in cancer metastases. Cold Spring Harb Perspect Med 2020; 10( 10): a036848

DOI

93
Wang M, Xia F, Wei Y, Wei X. Molecular mechanisms and clinical management of cancer bone metastasis. Bone Res 2020; 8( 1): 30

DOI

94
Weilbaecher KN, Guise TA, McCauley LK. Cancer to bone: a fatal attraction. Nat Rev Cancer 2011; 11( 6): 411– 425

DOI

95
Clézardin P, Coleman R, Puppo M, Ottewell P, Bonnelye E, Paycha F, Confavreux CB, Holen I. Bone metastasis: mechanisms, therapies, and biomarkers. Physiol Rev 2021; 101( 3): 797– 855

DOI

96
Bakir B, Chiarella AM, Pitarresi JR, Rustgi AK. EMT, MET, plasticity, and tumor metastasis. Trends Cell Biol 2020; 30( 10): 764– 776

DOI

97
Mohme M, Riethdorf S, Pantel K. Circulating and disseminated tumour cells—mechanisms of immune surveillance and escape. Nat Rev Clin Oncol 2017; 14( 3): 155– 167

DOI

98
Fidler IJ, Nicolson GL. Fate of recirculating B16 melanoma metastatic variant cells in parabiotic syngeneic recipients. J Natl Cancer Inst 1977; 58( 6): 1867– 1872

DOI

99
Liotta LA, Vembu D, Saini RK, Boone C. In vivo monitoring of the death rate of artificial murine pulmonary micrometastases. Cancer Res 1978; 38( 5): 1231– 1236

100
Fidler IJ. Metastasis: quantitative analysis of distribution and fate of tumor emboli labeled with 125I-5-iodo-2′-deoxyuridine. J Natl Cancer Inst 1970; 45( 4): 773– 782

101
Kim MY, Oskarsson T, Acharyya S, Nguyen DX, Zhang XH, Norton L, Massagué J. Tumor self-seeding by circulating cancer cells. Cell 2009; 139( 7): 1315– 1326

DOI

102
Zhang W, Bado IL, Hu J, Wan YW, Wu L, Wang H, Gao Y, Jeong HH, Xu Z, Hao X, Lege BM, Al-Ouran R, Li L, Li J, Yu L, Singh S, Lo HC, Niu M, Liu J, Jiang W, Li Y, Wong STC, Cheng C, Liu Z, Zhang XH. The bone microenvironment invigorates metastatic seeds for further dissemination. Cell 2021; 184( 9): 2471– 2486.e20

DOI

103
Mukai R, Tomimaru Y, Nagano H, Eguchi H, Mimori K, Tomokuni A, Asaoka T, Wada H, Kawamoto K, Marubashi S, Doki Y, Mori M. miR-615-3p expression level in bone marrow is associated with tumor recurrence in hepatocellular carcinoma. Mol Clin Oncol 2015; 3( 3): 487– 494

DOI

104
Sugimachi K, Sakimura S, Tomokuni A, Uchi R, Hirata H, Komatsu H, Shinden Y, Iguchi T, Eguchi H, Masuda T, Morita K, Shirabe K, Eguchi H, Maehara Y, Mori M, Mimori K. Identification of recurrence-related microRNAs from bone marrow in hepatocellular carcinoma patients. J Clin Med 2015; 4( 8): 1600– 1611

DOI

105
Deng L, Wang C, He C, Chen L. Bone mesenchymal stem cells derived extracellular vesicles promote TRAIL-related apoptosis of hepatocellular carcinoma cells via the delivery of microRNA-20a-3p. Cancer Biomark 2021; 30( 2): 223– 235

DOI

106
Zhang XH, Jin X, Malladi S, Zou Y, Wen YH, Brogi E, Smid M, Foekens JA, Massagué J. Selection of bone metastasis seeds by mesenchymal signals in the primary tumor stroma. Cell 2013; 154( 5): 1060– 1073

DOI

107
Hemingway F, Taylor R, Knowles HJ, Athanasou NA. RANKL-independent human osteoclast formation with APRIL, BAFF, NGF, IGF I and IGF II. Bone 2011; 48( 4): 938– 944

DOI

108
Andrade K, Fornetti J, Zhao L, Miller SC, Randall RL, Anderson N, Waltz SE, McHale M, Welm AL. RON kinase: a target for treatment of cancer-induced bone destruction and osteoporosis. Sci Transl Med 2017; 9( 374): eaai9338

DOI

109
D’Amico L, Roato I. The impact of immune system in regulating bone metastasis formation by osteotropic tumors. J Immunol Res 2015; 2015 : 143526

DOI

110
Muscarella AM, Aguirre S, Hao X, Waldvogel SM, Zhang XH. Exploiting bone niches: progression of disseminated tumor cells to metastasis. J Clin Invest 2021; 131( 6): e143764

DOI

111
Wu J, Lanier LL. Natural killer cells and cancer. Adv Cancer Res 2003; 90 : 127– 156

DOI

112
Okamoto T, Yoneyama MS, Hatakeyama S, Mori K, Yamamoto H, Koie T, Saitoh H, Yamaya K, Funyu T, Fukuda M, Ohyama C, Tsuboi S. Core2 O-glycan-expressing prostate cancer cells are resistant to NK cell immunity. Mol Med Rep 2013; 7( 2): 359– 364

DOI

113
Lo CH, Lynch CC. Multifaceted roles for macrophages in prostate cancer skeletal metastasis. Front Endocrinol (Lausanne) 2018; 9 : 247

DOI

114
Soki FN, Cho SW, Kim YW, Jones JD, Park SI, Koh AJ, Entezami P, Daignault-Newton S, Pienta KJ, Roca H, McCauley LK. Bone marrow macrophages support prostate cancer growth in bone. Oncotarget 2015; 6( 34): 35782– 35796

DOI

115
Lu X, Kang Y. Chemokine (C-C motif) ligand 2 engages CCR2+ stromal cells of monocytic origin to promote breast cancer metastasis to lung and bone. J Biol Chem 2009; 284( 42): 29087– 29096

DOI

116
Zhao E, Wang L, Dai J, Kryczek I, Wei S, Vatan L, Altuwaijri S, Sparwasser T, Wang G, Keller ET, Zou W. Regulatory T cells in the bone marrow microenvironment in patients with prostate cancer. OncoImmunology 2012; 1( 2): 152– 161

DOI

117
Yoldi G, Pellegrini P, Trinidad EM, Cordero A, Gomez-Miragaya J, Serra-Musach J, Dougall WC, Muñoz P, Pujana MA, Planelles L, González-Suárez E. RANK signaling blockade reduces breast cancer recurrence by inducing tumor cell differentiation. Cancer Res 2016; 76( 19): 5857– 5869

DOI

118
Tan W, Zhang W, Strasner A, Grivennikov S, Cheng JQ, Hoffman RM, Karin M. Tumour-infiltrating regulatory T cells stimulate mammary cancer metastasis through RANKL-RANK signalling. Nature 2011; 470( 7335): 548– 553

DOI

119
Ahern E, Smyth MJ, Dougall WC, Teng MWL. Roles of the RANKL-RANK axis in antitumour immunity—implications for therapy. Nat Rev Clin Oncol 2018; 15( 11): 676– 693

DOI

120
Shiozawa Y, Pedersen EA, Havens AM, Jung Y, Mishra A, Joseph J, Kim JK, Patel LR, Ying C, Ziegler AM, Pienta MJ, Song J, Wang J, Loberg RD, Krebsbach PH, Pienta KJ, Taichman RS. Human prostate cancer metastases target the hematopoietic stem cell niche to establish footholds in mouse bone marrow. J Clin Invest 2011; 121( 4): 1298– 1312

DOI

121
Okamoto K, Takayanagi H. Regulation of bone by the adaptive immune system in arthritis. Arthritis Res Ther 2011; 13( 3): 219

DOI

122
Gagliani N, Amezcua Vesely MC, Iseppon A, Brockmann L, Xu H, Palm NW, de Zoete MR, Licona-Limón P, Paiva RS, Ching T, Weaver C, Zi X, Pan X, Fan R, Garmire LX, Cotton MJ, Drier Y, Bernstein B, Geginat J, Stockinger B, Esplugues E, Huber S, Flavell RA. Th17 cells transdifferentiate into regulatory T cells during resolution of inflammation. Nature 2015; 523( 7559): 221– 225

DOI

123
Almand B, Clark JI, Nikitina E, van Beynen J, English NR, Knight SC, Carbone DP, Gabrilovich DI. Increased production of immature myeloid cells in cancer patients: a mechanism of immunosuppression in cancer. J Immunol 2001; 166( 1): 678– 689

DOI

124
Bronte V, Brandau S, Chen SH, Colombo MP, Frey AB, Greten TF, Mandruzzato S, Murray PJ, Ochoa A, Ostrand-Rosenberg S, Rodriguez PC, Sica A, Umansky V, Vonderheide RH, Gabrilovich DI. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun 2016; 7( 1): 12150

DOI

125
Shurin MR, Naiditch H, Zhong H, Shurin GV. Regulatory dendritic cells: new targets for cancer immunotherapy. Cancer Biol Ther 2011; 11( 11): 988– 992

DOI

126
Capietto AH, Faccio R. Immune regulation of bone metastasis. Bonekey Rep 2014; 3 : 600

DOI

127
Sawant A, Hensel JA, Chanda D, Harris BA, Siegal GP, Maheshwari A, Ponnazhagan S. Depletion of plasmacytoid dendritic cells inhibits tumor growth and prevents bone metastasis of breast cancer cells. J Immunol 2012; 189( 9): 4258– 4265

DOI

128
Esen E, Long F. Aerobic glycolysis in osteoblasts. Curr Osteoporos Rep 2014; 12( 4): 433– 438

DOI

129
Le A, Cooper CR, Gouw AM, Dinavahi R, Maitra A, Deck LM, Royer RE, Vander Jagt DL, Semenza GL, Dang CV. Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression. Proc Natl Acad Sci USA 2010; 107( 5): 2037– 2042

DOI

130
Park S, Chang CY, Safi R, Liu X, Baldi R, Jasper JS, Anderson GR, Liu T, Rathmell JC, Dewhirst MW, Wood KC, Locasale JW, McDonnell DP. ERRα-regulated lactate metabolism contributes to resistance to targeted therapies in breast cancer. Cell Rep 2016; 15( 2): 323– 335

DOI

131
Angelin A, Gil-de-Gómez L, Dahiya S, Jiao J, Guo L, Levine MH, Wang Z, Quinn WJ 3rd, Kopinski PK, Wang L, Akimova T, Liu Y, Bhatti TR, Han R, Laskin BL, Baur JA, Blair IA, Wallace DC, Hancock WW, Beier UH. Foxp3 reprograms T cell metabolism to function in low-glucose, high-lactate environments. Cell Metab 2017; 25( 6): 1282– 1293.e7

DOI

132
Ell B, Kang Y. SnapShot: bone metastasis. Cell 2012; 151( 3): 690– 690.e1

DOI

133
Huang Z, Chu L, Liang J, Tan X, Wang Y, Wen J, Chen J, Wu Y, Liu S, Liao J, Hou R, Ding Z, Zhang Z, Liang H, Song S, Yang C, Zhang J, Guo T, Chen X, Zhang B. H19 promotes HCC bone metastasis through reducing osteoprotegerin expression in a protein phosphatase 1 catalytic subunit alpha/p38 mitogen-activated protein kinase-dependent manner and sponging microRNA 200b-3p. Hepatology. 2021; 74( 1): 214– 232

DOI

134
Zhang S, Xu Y, Xie C, Ren L, Wu G, Yang M, Wu X, Tang M, Hu Y, Li Z, Yu R, Liao X, Mo S, Wu J, Li M, Song E, Qi Y, Song L, Li J. RNF219/α-catenin/LGALS3 axis promotes hepatocellular carcinoma bone metastasis and associated skeletal complications. Adv Sci (Weinh) 2021; 8( 4): 2001961

DOI

135
Zhang L, Niu H, Ma J, Yuan BY, Chen YH, Zhuang Y, Chen GW, Zeng ZC, Xiang ZL. The molecular mechanism of lncRNA34a-mediated regulation of bone metastasis in hepatocellular carcinoma. Mol Cancer 2019; 18( 1): 120

DOI

136
Huang J, Hu W, Lin X, Wang X, Jin K. FRZB up-regulated in hepatocellular carcinoma bone metastasis. Int J Clin Exp Pathol 2015; 8( 10): 13353– 13359

137
Xiang ZL, Zeng ZC, Tang ZY, Fan J, He J, Zeng HY, Zhu XD. Potential prognostic biomarkers for bone metastasis from hepatocellular carcinoma. Oncologist 2011; 16( 7): 1028– 1039

DOI

138
Jin K, Lan H, Wang X, Lv J. Genetic heterogeneity in hepatocellular carcinoma and paired bone metastasis revealed by next-generation sequencing. Int J Clin Exp Pathol 2017; 10( 10): 10495– 10504

139
Xiang ZL, Zeng ZC, Fan J, Wu WZ, He J, Zeng HY, Tang ZY. A clinicopathological model to predict bone metastasis in hepatocellular carcinoma. J Cancer Res Clin Oncol 2011; 137( 12): 1791– 1797

DOI

140
Hou R, Wang YW, Liang HF, Zhang ZG, Liu ZM, Zhang BH, Zhang BX, Chen XP. Animal and cellular models of hepatocellular carcinoma bone metastasis: establishment and characterisation. J Cancer Res Clin Oncol 2015; 141( 11): 1931– 1943

DOI

141
Xiang ZL, Zeng ZC, Tang ZY, Fan J, Zhuang PY, Liang Y, Tan YS, He J. Chemokine receptor CXCR4 expression in hepatocellular carcinoma patients increases the risk of bone metastases and poor survival. BMC Cancer 2009; 9( 1): 176

DOI

142
Wang X, Lan H, Shen T, Gu P, Guo F, Lin X, Jin K. Perineural invasion: a potential reason of hepatocellular carcinoma bone metastasis. Int J Clin Exp Med 2015; 8( 4): 5839– 5846

143
Xiang ZL, Zhao XM, Zhang L, Yang P, Fan J, Tang ZY, Zeng ZC. MicroRNA-34a expression levels in serum and intratumoral tissue can predict bone metastasis in patients with hepatocellular carcinoma. Oncotarget 2016; 7( 52): 87246– 87256

DOI

144
Zhang L, Niu H, Yang P, Ma J, Yuan BY, Zeng ZC, Xiang ZL. Serum lnc34a is a potential prediction biomarker for bone metastasis in hepatocellular carcinoma patients. BMC Cancer 2021; 21( 1): 161

DOI

145
Ma ZJ, Wang Y, Li HF, Liu MH, Bi FR, Ma L, Ma H, Yan HL. LncZEB1-AS1 regulates hepatocellular carcinoma bone metastasis via regulation of the miR-302b-EGFR-PI3K-AKT axis. J Cancer 2020; 11( 17): 5118– 5128

DOI

146
Liu J, Chen S, Wang W, Ning BF, Chen F, Shen W, Ding J, Chen W, Xie WF, Zhang X. Cancer-associated fibroblasts promote hepatocellular carcinoma metastasis through chemokine-activated hedgehog and TGF-β pathways. Cancer Lett 2016; 379( 1): 49– 59

DOI

147
Sun C, Hu A, Wang S, Tian B, Jiang L, Liang Y, Wang H, Dong J. ADAM17-regulated CX3CL1 expression produced by bone marrow endothelial cells promotes spinal metastasis from hepatocellular carcinoma. Int J Oncol 2020; 57( 1): 249– 263

DOI

148
Campbell JP, Merkel AR, Masood-Campbell SK, Elefteriou F, Sterling JA. Models of bone metastasis. J Vis Exp 2012; ( 67): e4260

DOI

149
Shevrin DH, Kukreja SC, Ghosh L, Lad TE. Development of skeletal metastasis by human prostate cancer in athymic nude mice. Clin Exp Metastasis 1988; 6( 5): 401– 409

DOI

150
Li M, Zhou M, Gong M, Ma J, Pei F, Beamer WG, Shultz LD, Hock JM, Yu X. A novel animal model for bone metastasis in human lung cancer. Oncol Lett 2012; 3( 4): 802– 806

151
Tian Z, Wu L, Yu C, Chen Y, Xu Z, Bado I, Loredo A, Wang L, Wang H, Wu KL, Zhang W, Zhang XH, Xiao H. Harnessing the power of antibodies to fight bone metastasis. Sci Adv 2021; 7( 26): eabf2051

DOI

152
Yu C, Wang H, Muscarella A, Goldstein A, Zeng HC, Bae Y, Lee BH, Zhang XH. Intra-iliac artery injection for efficient and selective modeling of microscopic bone metastasis. J Vis Exp 2016; ( 115): e53982

DOI

153
Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordón-Cardo C, Guise TA, Massagué J. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 2003; 3( 6): 537– 549

DOI

154
Lelekakis M, Moseley JM, Martin TJ, Hards D, Williams E, Ho P, Lowen D, Javni J, Miller FR, Slavin J, Anderson RL. A novel orthotopic model of breast cancer metastasis to bone. Clin Exp Metastasis 1999; 17( 2): 163– 170

DOI

155
Broutier L, Mastrogiovanni G, Verstegen MM, Francies HE, Gavarró LM, Bradshaw CR, Allen GE, Arnes-Benito R, Sidorova O, Gaspersz MP, Georgakopoulos N, Koo BK, Dietmann S, Davies SE, Praseedom RK, Lieshout R, IJzermans JNM, Wigmore SJ, Saeb-Parsy K, Garnett MJ, van der Laan LJ, Huch M. Human primary liver cancer-derived organoid cultures for disease modeling and drug screening. Nat Med 2017; 23( 12): 1424– 1435

DOI

156
Lee S, Burner DN, Mendoza TR, Muldong MT, Arreola C, Wu CN, Cacalano NA, Kulidjian AA, Kane CJ, Jamieson CAM. Establishment and analysis of three-dimensional (3D) organoids derived from patient prostate cancer bone metastasis specimens and their xenografts. J Vis Exp 2020; ( 156): e60367

DOI

157
Nemeth JA, Harb JF, Barroso U Jr, He Z, Grignon DJ, Cher ML. Severe combined immunodeficient-hu model of human prostate cancer metastasis to human bone. Cancer Res 1999; 59( 8): 1987– 1993

158
Kuperwasser C, Dessain S, Bierbaum BE, Garnet D, Sperandio K, Gauvin GP, Naber SP, Weinberg RA, Rosenblatt M. A mouse model of human breast cancer metastasis to human bone. Cancer Res 2005; 65( 14): 6130– 6138

DOI

159
Shultz LD, Brehm MA, Garcia-Martinez JV, Greiner DL. Humanized mice for immune system investigation: progress, promise and challenges. Nat Rev Immunol 2012; 12( 11): 786– 798

DOI

160
Shultz LD, Ishikawa F, Greiner DL. Humanized mice in translational biomedical research. Nat Rev Immunol 2007; 7( 2): 118– 130

DOI

161
Li YH, Lv MF, Lu MS, Bi JP. Bone marrow mesenchymal stem cell-derived exosomal miR-338-3p represses progression of hepatocellular carcinoma by targeting ETS1. J Biol Regul Homeost Agents 2021; 35( 2): 617– 627

162
Baccin C, Al-Sabah J, Velten L, Helbling PM, Grünschläger F, Hernández-Malmierca P, Nombela-Arrieta C, Steinmetz LM, Trumpp A, Haas S. Combined single-cell and spatial transcriptomics reveal the molecular, cellular and spatial bone marrow niche organization. Nat Cell Biol 2020; 22( 1): 38– 48

DOI

163
Adjei IM, Sharma B, Peetla C, Labhasetwar V. Inhibition of bone loss with surface-modulated, drug-loaded nanoparticles in an intraosseous model of prostate cancer. J Control Release 2016; 232 : 83– 92

DOI

164
Mu CF, Shen J, Liang J, Zheng HS, Xiong Y, Wei YH, Li F. Targeted drug delivery for tumor therapy inside the bone marrow. Biomaterials 2018; 155 : 191– 202

DOI

165
Mbese Z, Aderibigbe BA. Bisphosphonate-based conjugates and derivatives as potential therapeutic agents in osteoporosis, bone cancer and metastatic bone cancer. Int J Mol Sci 2021; 22( 13): 6869

DOI

166
Sun W, Ge K, Jin Y, Han Y, Zhang H, Zhou G, Yang X, Liu D, Liu H, Liang XJ, Zhang J. Bone-targeted nanoplatform combining zoledronate and photothermal therapy to treat breast cancer bone metastasis. ACS Nano 2019; 13( 7): 7556– 7567

DOI

167
Herrmann IK, Wood MJA, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nat Nanotechnol 2021; 16( 7): 748– 759

DOI

168
Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di Giannatale A, Ceder S, Singh S, Williams C, Soplop N, Uryu K, Pharmer L, King T, Bojmar L, Davies AE, Ararso Y, Zhang T, Zhang H, Hernandez J, Weiss JM, Dumont-Cole VD, Kramer K, Wexler LH, Narendran A, Schwartz GK, Healey JH, Sandstrom P, Labori KJ, Kure EH, Grandgenett PM, Hollingsworth MA, de Sousa M, Kaur S, Jain M, Mallya K, Batra SK, Jarnagin WR, Brady MS, Fodstad O, Muller V, Pantel K, Minn AJ, Bissell MJ, Garcia BA, Kang Y, Rajasekhar VK, Ghajar CM, Matei I, Peinado H, Bromberg J, Lyden D. Tumour exosome integrins determine organotropic metastasis. Nature 2015; 527( 7578): 329– 335

DOI

Outlines

/