Introduction

The musculoskeletal system comprises the bones, cartilage, muscles, tendons, and ligaments, playing a critical role in providing structural support and coordinating movements within the human body. This complex system possesses intrinsic regenerative capabilities, however, its reparative capacity is subject to inherent limitations, especially concerning the regeneration of cartilage, tendons, and ligaments [1]. Pathological conditions, including trauma, infections, autoimmune diseases, and the physiological aging process, can induce irreversible damage to the musculoskeletal system, thus culminating in musculoskeletal disorders (MSDs) [54]. By applying external force to compress the SMPU/Mg scaffold above the transition temperature and temporarily fixing the shape below the transition temperature, they obtained small volumes of SMPU/Mg scaffold, which allowed the scaffold to be implanted into cranial defects with minimal wounds (Fig. 4A). Under NIR irradiation, the scaffolds gradually warmed up due to the photothermal effect, recovering their original shape to match the shape of the bone defect when the temperature exceeded the transition temperature (Fig. 4B-E). This process accelerated cranial defect repair through the release of Mg2+ (Fig. 4F). In another study, Wang et al. employed cryogenic four-dimensional (4D) printing to create a hierarchical porous nanocomposite scaffold comprising BP nanosheets, osteogenic peptides, and β-tricalcium phosphate/poly (lactic acid-co-trimethylene carbonate) for irregular bone defects based on computed tomography (CT) scan data [58]. Under photothermal conditions at 45°C, the scaffold’s shape was restored to match the irregular bone defects, facilitating bone repair through the release of osteogenic peptides.

Fig. 4
figure 4

Photothermal-responsive SMPU/Mg scaffolds for bone repair. A Schematic diagram of preparation of small volume SMPU/Mg scaffolds and recovery of their shape. B The process of shape recovery of SMPU/Mg scaffolds under NIR (808 nm, 1 W/cm2) irradiation. OS, original shape; TS-1, Temporary shape-1; TS-2, Temporary shape-2; RS, recovered shape. C SMPU/Mg scaffold shape recovery induced by NIR laser under 100g weight compression. Scale bar: 50 μm. D Schematic illustration of in vivo application of SMPU/Mg scaffolds. E The process of implantation of SMPU/Mg scaffolds and shape recovery under NIR irradiation. Scale bar: 5mm. F Equivalent stress distribution by finite element analysis of SMPU/Mg scaffolds indicate tight contact of the scaffold with the skull. Reproduced with permission [54]. Copyright 2022, Elsevier

Despite the promising potential of photothermal-responsive shape memory polymer scaffolds for bone repair applications, several considerations must be addressed in future studies. First, selecting an appropriate transformation temperature is pivotal. Transition temperatures below body temperature may cause the materials to recover their shape prematurely during implantation, whereas excessively high transition temperatures could lead to tissue damage around the scaffolds or inactivation of growth factors loaded within the materials. In addition, many shape memory polymers exhibit limited degradation ability, necessitating research efforts to adapt material degradation to the physiological process of bone repair. This can be achieved by carefully choosing suitable SMP materials or modifying existing SMP materials to enhance their degradation properties.

Bone Infection

Open bone injuries and the application of orthopedic implants pose an increased risk of bacterial invasion and colonization [65]. Upon infection of the bone, bacteria stimulate inflammatory cells to secrete various inflammatory factors, upregulate osteoblast RANKL/OPG ratios, promote osteoclast formation, and induce osteoclast apoptosis, resulting in an imbalance between bone resorption and bone formation and impaired bone repair [66]. Globally, the reported annual incidence of osteomyelitis is 21.8 out of hundred thousand people, and this figure continues to rise each year [67]. Unfortunately, due to specific pathogenic factors such as intracellular infection, osteocyte lacuno-canalicular network (OLCN) invasion, biofilm formation, and abscess formation, eradicating bacteria often proves challenging, leading to recurrence of bone infection in approximately 17% of patients [68, 69]. PDT and PTT, as non-contact, broad-spectrum and highly effective anti-infection techniques, have shown great potential in the treatment of bone infections. Here, we summarize the mechanisms associated with PDT- and PTT-based phototherapy techniques in the treatment of infected bone defects (Fig. 5).

Fig. 5
figure 5

Phototherapy techniques for the treatment of bone infections. Jablonski diagrams of the photophysical and photochemical basis of PDT A and PTT B. C Mechanisms of PTT- and PDT-based techniques for the treatment of infected bone defects

Antibacterial PDT and PTT

Antibacterial PDT is mainly mediated by the generation of hydroxyl radicals (•OH), singlet oxygen (1O2) and superoxide anions (O2•−) catalyzed by photosensitizers upon light excitation. These ROS induce oxidative damage to polysaccharides, lipids, proteins, and nucleic acids, leading to cell membrane disruption, DNA damage, and enzyme inactivation, ultimately causing bacterial death [9]. The multifaceted antibacterial mechanisms of ROS confer PDT with a broader antibacterial spectrum and reduced susceptibility to resistance compared to antibiotics, making it highly effective in the treatment of bone infections. In one study, the photosensitizer toluidine blue was used in PDT for osteomyelitis in rats, resulting in more than 99% reduction in bacterial load after 30 days of treatment, without bone resorption or microabscesses observed [70].

In addition to provide a favorable anti-infection microenvironment, bone repair can also be actively promoted by incorporating osteogenic drugs [71]. Yuan et al. introduced carbon-ZnO into PLLA, imparting the scaffold not only with excellent antibacterial properties via PDT but also promoting the formation of mineralized nodules in human osteoblast-like MG-63 cells through the release of Zn2+ [72]. Recently, **e et al. designed a self-assembled bilayer hydrogel with spatiotemporal modulation of the immune microenvironment for osteomyelitis treatment [48]. The upper layer of this hydrogel was an AC10A hydrogel loaded with Ag2S QDs@DSPE-mPEG2000-Ce6/Aptamer (AD-Ce6/Apt), while the lower layer was an AC10ARGD hydrogel loaded with MSCs. After implantation, the upper hydrogel first released AD-Ce6/Apt, which exhibited direct antibacterial effects via ROS generated by PDT and enhanced the antibacterial effect by recruiting and inducing macrophage M1 polarization via the generated ROS in the first 3-4 days. After this, the lower hydrogel released MSCs to promote macrophage M2 polarization for anti-inflammation and induced MSCs osteogenic differentiation for fracture healing.

Photothermal materials exert broad-spectrum antibacterial effects through photothermal conversion under laser irradiation, raising the local temperature to induce bacterial membrane rupture, protein denaturation, and DNA damage [9]. While bacteria can develop mechanisms such as reducing uptake, inactivating, and promoting efflux of anti-microbial substances, they are unable to develop resistance to heat generated by PTT. Combining antibacterial PTT with pro-bone repair drugs represents an effective strategy for treating infected bone defects. For instance, Yuan et al. constructed a composite coating of BP and hydroxyapatite (BPs@HA) on the implant surface, which employed PTT at 50°C to eliminate bacteria, alleviate inflammation, and promote fracture healing through the bioactive coating (Fig. 6A-C) [73]. To prevent tissue damage from high temperature, Wu et al. proposed a "Sequential Photothermal Mediation" strategy for treating infected bone defects [74]. In this study, BP nanosheets with zinc sulfonate ligand (ZnL2) were integrated onto hydroxyapatite scaffolds (ZnL2-BPs@HAP). The PTT temperature was controlled at 47-50°C for the initial three days to eliminate bacteria and reduce inflammatory cell infiltration. Subsequently, the PTT temperature was lowered to 40-42°C to minimize tissue damage while accelerating bone regeneration through mild PTT and the release of Zn2+ and PO43-.

Fig. 6
figure 6

Photothermal bone scaffolds for the treatment of bone infections. A Schematic diagram of the mechanism of BPs@HA composite coating to promote fracture healing. B Representative photographs of the surgical site (the implant was inserted into the cancellous bone of the tibia just below the tibial plateau) and colonies of bacteria from the implant surface. C Micro-CT 3D reconstruction of femur fracture sites after treatment with wire (stainless Kirschner wire), HA-wire, and BPs@HA-wire. Reproduced with permission [73]. Copyright 2022, American Chemical Society. D Transwell migration assay of Schwann cells treated with GelMA, GB (GelMA loaded with BP), GBM hydrogel. Scale bar: 100 μm. E Immunostaining of PC12 cells cultured in Schwann cells conditioned medium on day 5 (GBM hydrogel effectively promoted neurite outgrowth). F Antibacterial capacity of different hydrogels with and without laser irradiation. Micro-CT 3D reconstruction images G and colony number H of S. aureus-infected skull defects treated with different hydrogels and periodic 808 nm (1 W/cm2) laser irradiation. I NGF and J CGRP immunofluorescence staining images of skull tissue. Scale bar: 500 μm (top), 200μm (bottom). Reproduced with permission [75]. Copyright 2023, Wiley-VCH

Combined therapy

Heat generated by PTT and ROS generated by PDT are the primary antibacterial pathways in phototherapy. However, it is essential to acknowledge that these therapeutic modalities can potentially be harmful to healthy tissues [13]. To mitigate these concerns and optimize treatment outcomes, researchers are exploring the development of multifunctional antibacterial bone implants that reduce reliance on monotherapy and minimize associated tissue damage. Currently, a range of studies is investigating the combination of PTT or PDT with chemotherapy and CDT, as well as incorporating physical structures to achieve synergistic antibacterial effects in the treatment of bone infections (Table 1).

Table 1 Phototherapy-based antibacterial bone scaffolds

Combining PDT and PTT has emerged as a highly effective strategy for treating bone infections. In this approach, bone implants generate heat through photothermal conversion under laser irradiation, which inhibits bacterial viability and increases bacterial cell membrane permeability, enhancing their susceptibility to ROS [9]. This enables fewer ROS and lower temperatures to eliminate infections, resulting in powerful antibacterial effects and less tissue damage. Recently, Li et al. prepared BMP-2 microsphere-coated BPs@PLGA scaffolds (BMP-2@BPs) for PTT and PDT of infected bone defects [44]. In vivo, mild PTT at 41°C, in synergy with PDT, killed over 99% of methicillin-resistant Staphylococcus aureus (MRSA). Furthermore, the combination of mild PTT and BMP-2 effectively promoted the proliferation and osteogenic differentiation of periosteal-derived stem cells, accelerating the formation of new bone in infected cranial defects. Notably, infections often lead to localized nerve fiber necrosis, hindering the repair of infected bone defects [69, 75]. To address this, **g et al. developed a photosensitive conductive GBM hydrogel, gelatin methacrylate (GelMA) hydrogel loaded with magnesium-modified black phosphorus [75]. GBM hydrogel promoted Schwann cell migration and neurite outgrowth of PC12 cells via NGF secreted by Schwann cells and the released Mg2+ from hydrogel (Fig. 6D, E). In addition, GBM hydrogel showed potent bactericidal effects against Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) via PTT and PDT (Fig. 6F). In vivo, GBM hydrogel effectively killed bacteria localized in infected skull defects under NIR irradiation, promoted NGF expression and the number of CGRP-positive nerve fibers (CGRP is a key mediator connecting nerve and bone regeneration), and accelerated the repair of S. aureus-infected skull defects (Fig. 6G-J).

Bacteria can exist in two forms: floating cells and microbial aggregates, with the latter known as biofilms, characterized by microbial aggregates embedded in extracellular polymeric substances composed of polysaccharides, proteins, extracellular DNA, and lipids [84]. Due to the physical barrier effect, biofilms exhibit enhanced resistance, up to 1000 times greater than planktonic bacteria, owing to biofilm heterogeneity, adaptive stress response, and the formation of persistent cells [85]. To disrupt biofilms, increasing the local temperature through PTT is a primary approach to increase penetration of antibacterial drugs. Yang et al. developed PLLA/ZIF-8@GO scaffolds that increased the local temperature to approximately 50°C through photothermal conversion, enhancing the penetration of the antibacterial agent Zn2+ and achieving highly efficient antibacterial properties [79]. Similarly, Yuan et al. constructed a composite coating containing mesoporous polydopamine (MPDA), indocyanine green (ICG), and arginylglycylaspartic acid (RGD) peptides on Ti surface to obtain Ti-M/I/RGD implant [86]. This innovative design facilitated the penetration of ICG into the biofilm and increased the permeability of bacterial cell membranes via PTT. Subsequently, ROS were generated via PDT, effectively disrupting biofilm integrity and eradicating bacteria.

Bone tumors

Bone tumors encompass primary tumors and metastatic bone tumors [7]. Among them, osteosarcoma is the most common primary solid malignant bone tumor, traditionally treated with complete surgical resection of the tumor and chemotherapy. However, despite the introduction of chemotherapy, the long-term survival rates of 65%-70% remain unsatisfactory, and chemotherapy inevitably causes serious side effects such as nausea, hair loss, and bone marrow suppression [87]. Bone metastasis, a common complication of various cancers, is typically incurable and leads to pain and pathological fractures, significantly impacting patients' quality of life and survival [88]. Given the limitations of traditional treatment modalities, phototherapy has garnered increasing attention in the treatment of osteosarcoma and bone metastases due to its remote controllability, high spatial and temporal resolution, and minimally invasive nature [111]. Copyright 2018, Wiley-VCH. F Schematic diagram of the anti-tumor mechanism of CuP@PPy-ZOL NPs via synergistic PDT/PTT/CDT. G 3D micro-CT reconstruction images of metastatic tumor tibia at different angles in a mouse model of breast cancer bone metastasis after different treatments. H Schematic diagram of the interaction mechanism of MDA-MB-231 cells with RAW264.7 cells and MC3T3-E1 cells before and after treatment. I Scanning electron microscope images and energy dispersive X-ray elemental map** of calcium on the cranial surface of RAW264.7 cells cultured with skull bone for 7 days after different treatments. Reproduced with permission [116]. Copyright 2023, Wiley-VCH