Blog

  • A Novel Quadruple Conversion Therapy: Converting Initially Unresectabl

    A Novel Quadruple Conversion Therapy: Converting Initially Unresectabl

    Background

    Worldwide, hepatocellular carcinoma (HCC) is the sixth most frequently occurring cancer and the third leading cause of cancer mortality, accounting for 90% of primary liver malignancies.1 Despite advancements in therapeutic approaches, surgical removal continues to be the gold standard for curative treatment and provides the best potential for long-term survival.2 However, more than 70% of individuals with HCC are diagnosed at intermediate or advanced stages, where the majority have already lost the opportunity for direct surgical intervention.3,4 This limitation highlights the critical need for effective downstaging strategies to convert unresectable HCC to resectable disease. Successful conversion can not only significantly enhance the long-term outcomes of these patients but also lead to a complete pathological response (CPR) in certain patients who respond to conversion therapy, with survival outcomes comparable to those of patients initially considered to have resectable tumours.5,6

    As far back as the 1970s, reports indicated that individuals with HCC that was initially inoperable could achieve sufficient tumour downstaging to become eligible for surgical resection.7 As therapeutic agents and approaches for advanced HCC have progressed, treatment strategies have evolved, and multimodal, high-intensity approaches are attracting increasing attention, aiming to achieve rapid tumour shrinkage and downstaging or to augment the volume of the residual liver, ultimately providing an opportunity for curative surgical resection. Recent studies have indicated that locoregional therapy (LRT) combined with tyrosine kinase inhibitors (TKIs) and anti-programmed death-1 (PD-1) antibodies may serve as a superior conversion therapy for patients initially diagnosed with unresectable HCC while maintaining a manageable safety profile.8 Currently, the most frequently employed LRTs in clinical practice include TACE/TAE and HAIC. The ORR rate of a quadruple therapy combining TACE, HAIC, TKIs, and anti-PD-1 antibodies is 80%, theoretically suggesting a high potential conversion rate.9 However, no specific studies have been conducted on this topic, and this therapy is associated with considerable adverse effects, especially the potential deterioration of liver function following TACE. In TACE treatment, “complete embolization” is typically implied, but in recent years, a shift towards “partial embolization” has occurred.10,11 Partial transcatheter arterial embolization (pTAE) can prevent liver function deterioration caused by complete embolization while slowing blood flow, thereby increasing the local concentration and retention time of HAIC drugs. Additionally, pTAE induces the release of tumour antigens, thereby enhancing the antitumour effect of TKI combined with anti-PD-1 antibody treatment. This approach may also achieve a partial remission of intrahepatic lesions, potentially leading to a more effective conversion rate.10,11

    In this research, we assessed the potential of pTAE-HAIC in combination with TKIs and anti-PD-1 antibodies for downstaging along with resection in patients with HCC initially deemed unresectable We report a cohort of 17 patients with advanced or unresectable HCC who received pTAE-HAIC in combination with TKIs and anti-PD-1 antibodies and subsequently underwent successful R0 resection. To the best of our knowledge, this study represents the largest reported cohort of patients with unresectable HCC to achieve R0 resection after receiving this quadruple therapy approach.

    Patients and Methods

    A retrospective analysis was conducted on patients diagnosed with HCC at Chongqing University Cancer Hospital from September 1, 2021, to February 1, 2023, and treated with pTAE-HAIC plus TKI and anti-PD-1 antibody combination therapy. The diagnosis of HCC was based on both domestic and international guidelines.3,12 Patients with unresectable HCC are primarily categorized into two types: individuals with BCLC stages B‒C (advanced-stage cancer), who face a greater risk of resection failure or reduced survival than those who receive palliative treatment, or those with an insufficient remnant liver volume following hepatectomy (less than 40% in patients with cirrhosis; less than 30% in patients without cirrhosis). The exclusion criteria were as follows: (1) Child‒Pugh score of Grade C; (2) Eastern Cooperative Oncology Group (ECOG)/Preservation Scale (PS) score of 3–4; (3) a history of malignant tumours other than HCC; (4) the presence of extrahepatic metastasis; (5) serious cardiac, pulmonary, or renal insufficiency; and (6) incomplete follow-up data.

    The research protocol, encompassing the therapy schedule and methods for gathering data, adhered to the ethical guidelines outlined in the World Medical Association’s Declaration of Helsinki and was approved by the Research Ethics Committee of Chongqing University Cancer Hospital (Approval Number: CZLS2021042-A), and written informed consent was obtained from all patients prior to undergoing pTAE-HAIC, TKI, and anti-PD-1 antibody treatment and surgery.

    pTAE-HAIC Procedure

    A preoperative enhanced CT scan was performed to assess all the tumour-feeding arteries. During the procedure, all extrahepatic collateral arteries were first embolized, followed by the embolization of the major intrahepatic feeding arteries. In cases where hepatic artery-to-portal vein or hepatic artery-to-hepatic vein fistulas were present, embolization procedures should avoid the fistula site. If avoidance was not possible, appropriate embolic agents, such as particles or coils, should be used to effectively occlude the fistula. These measures should be implemented prior to embolizing the artery that supplies blood to the tumour to ensure the safety and efficacy of the treatment. The pTAE-HAIC treatment protocol and schematic are detailed in Figures 1 and 2. TAE utilized ≤10 mL of ultrafine iodized oil (purchased from Jiangsu Hengrui Medicine China Co., Ltd)., along with suitable particulate embolic materials (including embolic microspheres (purchased from Jiangsu Hengrui Medicine China Co., Ltd)., polyvinyl alcohol (PVA) particle embolic agents (purchased from Varian Medical Systems China Co., Ltd)., gelatine sponge particle embolic agents (purchased from Varian Medical Systems China Co., Ltd)., and coils (purchased from COOK Medical, USA)). Following TAE, a catheter was placed at the origin of the tumour-feeding artery for FOLFOX-HAIC (arterial infusion of oxaliplatin at a dose of 85 mg/m² for a duration of 2–3 hours; leucovorin at a dose of 400 mg/m² was administered through an arterial infusion over a period of 2 hours, whereas 5-FU at 2400 mg/m² was administered via a continuous arterial infusion for 46 hours). pTAE-HAIC treatment is administered once every 4–6 weeks.

    Figure 1 pTAE-HAIC treatment protocol.

    Figure 2 Schematic diagram of pTAE-HAIC, the grey areas denote the embolized vessels and tumor.

    Systemic Therapy

    This study utilized different treatment regimens according to local practices and research protocols.13,14 The TKIs used included lenvatinib15 (administered at a fixed dose of 8 mg/day, without considering the weight of the patient), apatinib16 (administered at 250 mg/day), and sorafenib17 (administered at 400 mg twice daily). The following anti-PD-1 antibodies were administered intravenously: tislelizumab18 at 200 mg every three weeks, camrelizumab19 at 200 mg every two weeks, or sintilimab20 at 200 mg every three weeks. All patients received regular treatments and follow-up monitoring. Specifically, comprehensive evaluations, including complete blood counts and assessments of cardiac, thyroid, adrenal, renal, and liver functions, along with oncological markers, were conducted every 2 to 3 weeks before each cycle of anti-PD-1 antibody therapy. Contrast-enhanced computed tomography (CT) or magnetic resonance imaging (MRI), as well as chest CT, were used to assess tumour response and resectability approximately every month (±1 week). The tumour response was assessed in accordance with RECIST v1.1 and the modified RECIST (mRECIST).21,22 The monitoring and grading of adverse events were conducted using the National Cancer Institute’s Common Terminology Criteria for Adverse Events version 4.0.

    Liver Resection Procedure

    The resectability of the tumour was determined from the imaging findings according to the following criteria:23 (1) achieving R0 resection with a sufficient remaining liver volume and function; (2) intrahepatic lesions were evaluated as a complete response (CR), partial response (PR), or stable disease (SD); (3) no serious or sustained adverse reactions were caused by any treatment; and (4) no contraindications for hepatectomy were present. Tumour resectability was discussed and approved by the multidisciplinary team (MDT) at Chongqing University Cancer Hospital, which included senior surgeons and radiologists.

    Posthepatectomy liver failure (PHLF) was diagnosed and graded according to the criteria established by the International Study Group of Liver Surgery (ISGLS). PHLF is defined as an elevated international normalized ratio (INR) of prothrombin time and hyperbilirubinemia occurring on or after the fifth postoperative day.24 Complications after surgery were categorized using the Clavien‒Dindo classification system.25 A pathological complete response (pCR) was characterized by the absence of viable residual tumour cells in haematoxylin- and eosin-stained sections of the primary tumour, tumour thrombi, and metastatic lesions that were completely resected.

    Targeted immunotherapy was resumed four weeks postoperatively, with follow-up imaging and serum tumour biomarker assessments conducted every 1–2 months.

    Statistical Analyses

    All the statistical analyses were conducted using R software, version 3.5.3. Overall survival (OS) was calculated as the duration from the initiation of combination therapy to the patient’s death. Recurrence-free survival (RFS) was determined as the interval from the date of the surgical intervention to the first occurrence of either tumour recurrence or death from any cause.

    Results

    Patients and Treatment

    Between October 2021 and February 2023, 62 consecutive patients who received pTAE-HAIC combined with TKIs and anti-PD-1 antibodies as the first-line therapy were included in the analysis (Figure 3). Among these patients, 17 (27.4%) successfully underwent R0 resection. An additional 3 patients were deemed to have resectable tumours following the achievement of the PR of the primary lesion, thereby meeting the first and second resection criteria mentioned above. However, these three patients did not undergo liver resection—one patient experienced persistent adverse effects from anti-PD-1 antibody therapy (grade II hypothyroidism and grade II myocarditis), while the remaining two patients declined surgery.

    Figure 3 Patient flowchart.

    Table 1 provides a summary of the demographics and baseline characteristics of all 62 patients who underwent quadruple conversion therapy. According to the classification by the Liver Cancer Study Group of Japan (LCSGJ),26 none of the 13 patients with portal vein trunk and/or superior mesenteric vein invasion classified as Vp4 underwent surgery after treatment (p = 0.031). Among the remaining 49 patients, 17 underwent surgery. Among these 17 patients, 13 were male and 4 were female, with a median age of 51 years (range: 18–70 years). Twelve patients had BCLC stage C disease, of whom one had an ECOG score of 1. Among these patients, 11 had major vascular invasion, including 4 patients classified as Vp2 and 4 as Vp3. One patient had hepatic vein invasion classified as Vv1, 3 patients were classified as Vv2, and 2 patients exhibited invasion of the inferior vena cava (IVC) and/or right atrium (RA) and were classified as Vv3. Among these patients, 3 had concurrent invasion of both the portal and hepatic venous systems, specifically, Vp2/Vv2, Vp3/Vv2, and Vp3/Vv3. Additionally, 5 patients had BCLC stage B HCC.

    Table 1 Baseline Patient Demographics and Disease Characteristics

    The median diameter of the largest hepatic nodule was 11.5 cm (range: 3.9–18.8 cm), and 10 patients had multiple liver nodules. Before surgery, the 17 patients who underwent successful surgical resection were administered 43 cycles of pTAE-HAIC (range: 1–5, median: 2). The tumour response was evaluated prior to hepatectomy via both RECIST v1.1 and mRECIST. The TKIs and anti-PD-1 antibodies administered to these 17 patients are listed in Table 2. In accordance with RECIST v1.1, 13 patients achieved a PR and 4 achieved SD. Using mRECIST, 3 patients with a CR, 12 patients with a PR, and 2 patients with SD were identified (Table 2).

    Table 2 Characteristics of Surgical and Postoperative Features

    Surgical and Perioperative Findings

    The median time from the initiation of quadruple therapy to surgery was 89 days (range: 69–255 days). Among patients with BCLC stage C disease, all 13 patients with Vp4-grade vascular invasion failed to achieve successful conversion (P=0.031), whereas 14 of 21 patients (66.7%) with vascular invasion achieved conversion to resectable disease, including two patients who attained an R0 resection status but did not undergo surgery. The vascular invasion grades of these two patients were Vp3 and Vv2, respectively (Table 1). All participants who underwent surgery had a preoperative Child‒Pugh classification of Grade A. As presented in Table 3, a total of 11 patients underwent major hepatectomy procedures (≥3 segments), with a median intraoperative blood loss volume of 300 mL (range: 100–1500 mL). The median duration of postoperative hospitalization was 13 days (range: 6–18 days). Three patients (17.6%) experienced liver failure after surgery, but their conditions improved following liver-protective treatment. Postoperative adverse events are listed in Table 4, with only two cases of grade III adverse events, both of which were peritonitis, which were resolved with antibiotic treatment.

    Table 3 Patient Characteristics Before and After Surgery

    Table 4 Postoperative Adverse Events

    Seven patients (41%) achieved pCR across all surgical specimens. In Patient 11, the resected middle hepatic vein tumour thrombus achieved pCR, while a few viable tumour cells were still present in the intrahepatic lesions, resulting in a downstaging of the BCLC classification from Stage C to Stage A. Additionally, in Patient 15, the intrahepatic tumour achieved pCR, but a few viable tumour cells were still observed in the right anterior branch of the PVTT.

    Follow-Up

    All patients continued receiving systemic therapy and regular follow-up after surgery. Among the 17 patients, with a median observation period of 17.8 months (range: 12.2–38.3 months), the 12-month OS rate after hepatectomy was 100%, and the median postoperative PFS was 14.5 months (range: 1.5–31.8 months). Five patients experienced tumour recurrence within 12 months, with only one patient experiencing continued progression, and the remaining four patients achieved disease control following treatment. Patients 3 and 6 developed pulmonary metastasis; after changing the systemic treatment regimen, Patient 3 achieved a stable disease, with no new tumours detected at the last follow-up, whereas Patient 6 subsequently developed new hepatic, bone, and lymph node metastases. Patient 5 experienced recurrence at the hepatic surgical site and underwent repeat partial hepatectomy, with no recurrence observed at the last follow-up. Patients 9 and 11 developed intrahepatic metastasis postoperatively, which was well controlled with interventional therapy, with no recurrence noted at the final follow-up.

    Treatment Safety

    The adverse events are detailed in Table 4. Among the grade 3 adverse events, two cases (11.8%) of peritoneal infection were observed, both of which improved with antibiotic treatment, leading to discharge. The most common grade 1–2 adverse events included hypoalbuminaemia (9 patients, 52.9%), pleural effusion (7 patients, 41.2%), and hypokalaemia (6 patients, 35.3%).

    Discussion

    In this study, 62 patients with HCC that was initially inoperable received conversion therapy, and 27.4% (17/62) achieved R0 resection. Quadruple therapy consisting of pTAE-HAIC combined with TKIs and anti-PD-1 antibodies represents a feasible conversion strategy for patients with unresectable HCC to achieve successful resection and potential long-term survival.

    The quadruple combination therapy, which includes pTAE-HAIC, TKIs, and anti-PD-1 antibodies, has a high objective response rate (ORR), with significant short-term tumour reduction observed in our study (Figure 4a–d). One of the most critical factors for achieving successful conversion is the ORR. Quick effectiveness reduces the duration of exposure to conversion therapy, thus lowering the risk of negative side effects. A more rapid ORR indicates a greater likelihood of shrinking the tumour size and downgrading the stage, which is undoubtedly more favourable for subsequent surgical resection.14

    Figure 4 Four representative cases.

    Notes: Patient 7 (a) was diagnosed with multiple HCC tumours in the right hepatic segment, with invasion into the right anterior branch of the portal vein (indicated by the red arrow). The tumour stage was classified as BCLC stage C. The patient was administered 8 mg of lenvatinib daily and 200 mg of tislelizumab every 3 weeks combined with two cycles of pTAE-HAIC (as shown via angiography, only one main tumour-feeding artery was embolized, with the blue arrow indicating its location). The catheter was inserted and positioned within the right hepatic artery to facilitate HAIC. Following quadruple therapy, the tumours exhibited substantial shrinkage, and the primary tumours and portal vein tumour thrombosis showed no arterial enhancement on a contrast-enhanced CT scan. Curative liver resection was performed, and the patient transitioned from planned hemiliver resection to segmental resection following conversion therapy. The surgically removed specimen displayed a pCR upon H&E staining. Patient 10 (b) was identified with an isolated HCC tumour located in the right hepatic segment, with invasion into the right branch of the portal vein (indicated by the red arrow). The tumour was classified as BCLC stage C. The patient was administered 8 mg of lenvatinib daily and 200 mg of sintilimab every 3 weeks combined with four cycles of pTAE-HAIC (as shown by angiography, only two main tumour-feeding arteries were embolized, with the blue arrow indicating its location). The microcatheter was inserted and positioned within the right hepatic artery to facilitate HAIC. Following quadruple therapy, the tumour displayed substantial shrinkage, and the primary tumour and PVTT did not exhibit arterial enhancement on a contrast-enhanced CT scan and MRI. Curative liver resection was performed, and the patient transitioned from planned hemiliver resection to segmental resection following conversion therapy. The surgically removed specimen displayed a pCR upon H&E staining. Patient 11 (c) was found to have an isolated HCC tumour that invaded the middle hepatic vein (indicated by the red arrow). The tumour was classified as BCLC stage C and the patient had an insufficient remnant liver volume following hepatectomy. The patient was administered 8 mg of lenvatinib daily and 200 mg of tislelizumab every 3 weeks combined with four cycles of pTAE-HAIC (as shown via angiography, only two main tumour-feeding arteries were embolized, with the blue arrow indicating its location). The microcatheter was inserted and positioned within the proper hepatic artery to facilitate HAIC. Following quadruple therapy, the tumours displayed substantial shrinkage, and presurgery CT and MRI scans indicated the disappearance of the tumour thrombus in the middle hepatic vein, suggesting downstaging from BCLC stage C to stage B; moreover, the remnant liver volume increased significantly. Curative liver resection was performed. Patient 16 (d) was identified with a solitary HCC tumour located in the right hepatic segment, which invaded both the right anterior and posterior branches of the portal vein (indicated by the red arrow). The tumour was categorized as BCLC stage C. The patient was administered 8 mg of lenvatinib daily and 200 mg of tislelizumab every 3 weeks combined with four cycles of pTAE-HAIC (as shown by angiography, the lateral hepatic branch blood supply was embolized, with the right inferior phrenic artery, with the blue arrow indicating its location). The microcatheter was inserted and positioned within the proper hepatic artery to facilitate HAIC. All extrahepatic collateral arteries should be embolized first, as in this patient. Following successful quadruple therapy, curative liver resection was performed.

    Why do we consider and recommend pTAE rather than TACE in combination therapy? Typically, TACE refers to complete chemoembolization of all lesions within the liver, aiming to maximize the CR rate. The latest studies have shown that the combination of TACE-HAIC with targeted and immune therapies can provide better efficacy for patients with intermediate and advanced HCC along with PVTT.9,27 In these studies, TACE utilized epirubicin, whereas HAIC employed oxaliplatin, leucovorin, and fluorouracil. However, the combined use of multiple chemotherapeutic agents can exacerbate liver damage. This damage is further compounded by the liver function impairment attributable to complete embolization. According to previous reports, the incidence of liver damage in the combination therapy group was markedly higher than that in the TACE-alone control group (68.3% vs 47.6%).27 Poorer liver function may not only affect subsequent treatments but also lead to liver failure, ultimately shortening the survival period. Therefore, when using the quadruple combination therapy, we do not recommend the use of multiple chemotherapeutic agents during TACE but instead advocate simple TAE. In recent years, the role of TACE in combination therapies has also undergone significant transformation, gradually shifting from “complete embolization” to “partial embolization”. Our centre has been exploring the combination of pTAE and HAIC with targeted therapy and immunotherapy for quite some time, and this approach has been widely applied in clinical practice. We have also developed a procedural diagram and schematic diagram for pTAE-HAIC (Figures 1 and 2). Additionally, adverse reactions following successful surgical resection after conversion are controllable.

    In the quadruple combination therapy, pTAE plays a crucial synergistic role. pTAE effectively reduces the tumour burden while preventing liver function deterioration and postembolization syndrome associated with complete embolization. Since the HAIC catheter is typically placed within the hepatic artery, pTAE requires the embolization of all extrahepatic collateral arteries involved in the tumour blood supply; simultaneously, embolizing the major intrahepatic feeding arteries slows blood flow and increases the local drug concentration and exposure time, thereby increasing the efficacy of HAIC chemotherapy.28 Additionally, pTAE induces tumour cell death, leading to the release of antigens and modification of the tumour microenvironment, which enhance the immune response.10,11 Finally, when used in conjunction with TKIs, these agents can counteract the angiogenic activity induced by hypoxia following embolization.29 Overall, the multifaceted benefits of pTAE contribute to a more effective and well-tolerated therapeutic regimen in the quadruple combination therapy.

    Notably, none of the 13 patients with portal vein trunk/superior mesenteric vein invasion classified as Vp4 underwent surgery after treatment (P=0.031). Previous research has shown that although surgical resection may provide a more favourable prognosis for HCC patients who have PVTT than nonsurgical therapies, including TACE/HAIC or sorafenib,30 subgroup analyses have revealed that if the neoplastic thrombus has spread to the main trunk of the portal vein (Vp4), the prognosis following combined liver resection and thrombectomy does not seem to be superior to that of nonsurgical treatments.30,31 Moreover, Vp4 independently predicts a poor prognosis for HCC patients, and treatment outcomes for patients with PVTT Vp4 are generally suboptimal, whether through interventional therapy, targeted therapy, or a combination of targeted and immune therapy, with a limited survival benefit.32 Therefore, the conversion rate for Vp4 patients is likely to be the lowest, and the timing for surgery after conversion may need to be more cautious. Whether achieving a CR (complete response) in the main trunk of the PVTT according to mRECIST is the optimal surgical timing remains to be determined. This question warrants further investigation.

    This study has several limitations that warrant discussion. First, the postoperative follow-up duration was relatively short, precluding a comprehensive understanding of the long-term outcomes. Second, the combination of TKIs and anti-PD-1 antibodies used in this study was not uniform. Treatment decisions can evolve over time with the emergence of new clinical guidelines and research findings. Third, this analysis was conducted retrospectively at a single centre, which introduces the potential for selection bias. Well-designed prospective randomized controlled trials are needed in the future to further confirm these findings.

    In summary, the findings reveal that the novel quadruple conversion therapy strategy, which combines pTAE-HAIC with TKIs and anti-PD-1 antibodies, is feasible for treating initially unresectable HCC, subsequent liver resection is both effective and safe, demonstrating potential long-term survival benefits after successful conversion therapy. Nevertheless, large-scale prospective studies will be required to confirm the observed therapeutic advantages.

    Data Sharing Statement

    All data used during the study are available from the corresponding author RongZhong Huang, Rong Zhou upon reasonable request.

    Ethics Statement

    The research protocol, encompassing the therapy schedule and methods for gathering data, adhered to the ethical guidelines outlined in the World Medical Association’s Declaration of Helsinki and was approved by the Research Ethics Committee of Chongqing University Cancer Hospital (Approval Number: CZLS2021042-A), and written informed consent was obtained from all patients prior to undergoing pTAE-HAIC, TKI, and anti-PD-1 antibody treatment and surgery.

    Author Contributions

    All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

    Funding

    This study was supported by the Joint project of Chongqing Health Commission and Science and Technology Bureau (2020MSXM071).

    Disclosure

    The authors declare that there are no conflicts of interest.

    References

    1. Bray F, Laversanne M, Sung H, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74(3):229–263. doi:10.3322/caac.21834

    2. Reig M, Forner A, Rimola J, et al. BCLC strategy for prognosis prediction and treatment recommendation: the 2022 update. J Hepatol. 2022;76(3):681–693. doi:10.1016/j.jhep.2021.11.018

    3. Zhou J, Sun H, Wang Z, et al. Guidelines for the diagnosis and treatment of primary liver cancer (2022 edition). Liver Cancer. 2023;12(5):405–444. doi:10.1159/000530495

    4. Kloeckner R, Galle PR, Bruix J. Local and regional therapies for hepatocellular carcinoma. Hepatology. 2021;73 Suppl 1(S1):137–149. doi:10.1002/hep.31424

    5. Xu B, Zhu XD, Shen YH, et al. Criteria for identifying potentially resectable patients with initially oncologically unresectable hepatocellular carcinoma before treatment with lenvatinib plus an anti-PD-1 antibody. Front Immunol. 2022;13:1016736. doi:10.3389/fimmu.2022.1016736

    6. Wu SJ, Ruan DD, Wu QY, et al. Safety and efficacy of drug-eluting bead transarterial chemoembolization combined with lenvatinib and anti-PD-1 antibodies for unresectable hepatocellular carcinoma: a retrospective analysis. J Hepatocell Carcinoma. 2023;10:807–820. doi:10.2147/JHC.S408819

    7. Hermann RE, Lonsdale D. Chemotherapy, radiotherapy, and hepatic lobectomy for hepatoblastoma in an infant: report of a survival. Surgery. 1970;68(2):383–388. doi:10.1016/S0022-3468(70)80081-1

    8. Xu H, Zhang H, Li B, et al. Systemic conversion therapies for initially unresectable hepatocellular carcinoma: a systematic review and meta-analysis. BMC Cancer. 2024;24(1):1008. doi:10.1186/s12885-024-12772-y

    9. Li L, He J, Xie YX, et al. A retrospective controlled study of TACE-HAIC-targeted-immune quadruple therapy for intermediate and advanced-stage hepatocellular carcinoma. Zhonghua Gan Zang Bing Za Zhi. 2022;30(9):939–946. Chinese. doi:10.3760/cma.j.cn501113-20220823-00432

    10. Kudo M, Aoki T, Ueshima K, et al. Achievement of complete response and drug-free status by atezolizumab plus bevacizumab combined with or without curative conversion in patients with transarterial chemoembolization-unsuitable, intermediate-stage hepatocellular carcinoma: a multicenter proof-of-concept study. Liver Cancer. 2023;12(4):321–338. doi:10.1159/000529574

    11. Amashita T, Inaba Y, Ikeda M, et al. 205TiP IMPACT: randomized, multicenter, Phase III study evaluating the efficacy of immunotherapy (atezolizumab) plus anti-VEGF therapy (bevacizumab) in combination with transcatheter arterial chemoembolization for unresectable hepatocellular carcinoma (HCC). Ann Oncol. 2023;34:S1553–S1554. doi:10.1016/j.annonc.2023.10.784

    12. American Association for the Study of Liver Diseases; Bruix J, Sherman M. Management of hepatocellular carcinoma: an update. Hepatology. 2011;53(3):1020–1022. doi:10.1002/hep.24199

    13. Bureau of Medical Administration, National Health Commission of the People’s Republic of China. Standardization for diagnosis and treatment of hepatocellular carcinoma (2022 edition). Chinese J Hepatol. 2022;30(04):367–388. doi:10.3760/cma.j.cn501113-20220413-00193

    14. Alliance of Liver Cancer Conversion Therapy, Committee of Liver Cancer of the Chinese Anti-Cancer Association; Sun HC, Zhou J, Wang Z, et al. Chinese expert consensus on conversion therapy for hepatocellular carcinoma (2021 edition). Hepatobiliary Surg Nutr. 2022;11(2):227–252. doi:10.21037/hbsn-21-328

    15. Wang DX, Yang X, Lin JZ, et al. Efficacy and safety of lenvatinib for patients with advanced hepatocellular carcinoma: a retrospective, real-world study conducted in China. World J Gastroenterol. 2020;26(30):4465–4478. doi:10.3748/wjg.v26.i30.4465

    16. Qin S, Li Q, Gu S, et al. Apatinib as second-line or later therapy in patients with advanced hepatocellular carcinoma (AHELP): a multicentre, double-blind, randomised, placebo-controlled, Phase 3 trial. Lancet Gastroenterol Hepatol. 2021;6(7):559–568. doi:10.1016/S2468-1253(21)00109-6

    17. Park JW, Kim YJ, Kim DY, et al. Sorafenib with or without concurrent transarterial chemoembolization in patients with advanced hepatocellular carcinoma: the phase III STAH trial. J Hepatol. 2019;70(4):684–691. doi:10.1016/j.jhep.2018.11.029

    18. Qin S, Finn RS, Kudo M, et al. RATIONALE 301 study: tislelizumab versus sorafenib as first-line treatment for unresectable hepatocellular carcinoma. Future Oncol. 2019;15(16):1811–1822. doi:10.2217/fon-2019-0097

    19. Qin S, Ren Z, Meng Z, et al. Camrelizumab in patients with previously treated advanced hepatocellular carcinoma: a multicentre, open-label, parallel-group, randomised, Phase 2 trial. Lancet Oncol. 2020;21(4):571–580. doi:10.1016/S1470-2045(20)30011-5

    20. Zhang W, Bi X, Sun Y, et al. Preliminary results of sintilimab plus different dose of IBI305 (anti-VEGF monoclonal antibody) in patients with advanced hepatocellular carcinoma: a phase Ib study. J Clin Oncol. 2020;38(15_Suppll):3079. doi:10.1200/JCO.2020.38.15_suppl.3079

    21. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–247. doi:10.1016/j.ejca.2008.10.026

    22. Choi MH, Park GE, Oh SN, et al. Reproducibility of mRECIST in measurement and response assessment for hepatocellular carcinoma treated by transarterial chemoembolization. Acad Radiol. 2018;25(11):1363–1373. doi:10.1016/j.acra.2018.02.013

    23. Zhenyun CHENM. Discussion on case selection criteria for transformation therapy of hepatocellular carcinoma. Chinese J Oncol Prevention Treatm. 2021;13(5):464–468. doi:10.3969/j.issn.1674-5671.2021.05.04

    24. Rahbari NN, Garden OJ, Padbury R, et al. Posthepatectomy liver failure: a definition and grading by the international study group of liver surgery (ISGLS). Surgery. 2011;149(5):713–724. doi:10.1016/j.surg.2010.10.001

    25. Bolliger M, Kroehnert JA, Molineus F, et al. Experiences with the standardized classification of surgical complications (Clavien-Dindo) in general surgery patients. Eur Surg. 2018;50(6):256–261. doi:10.1007/s10353-018-0551-z

    26. HCC Expert Panel of Japan Society Of Hepatology; Kudo M, Izumi N, Kokudo N, et al. Management of hepatocellular carcinoma in Japan: consensus-based clinical practice guidelines proposed by the japan society of hepatology (JSH) 2010 updated version. Dig Dis. 2011;29(3):339–364. doi:10.1159/000327577

    27. Yuan Y, He W, Yang Z, et al. TACE-HAIC combined with targeted therapy and immunotherapy versus TACE alone for hepatocellular carcinoma with portal vein tumour thrombus: a propensity score matching study. Int J Surg. 2023;109(5):1222–1230. doi:10.1097/JS9.0000000000000256

    28. Zhao M, Guo Z, Zou YH, et al. Arterial chemotherapy for hepatocellular carcinoma in China: consensus recommendations. Hepatol Int. 2024;18(1):4–31. doi:10.1007/s12072-023-10599-6

    29. Cai M, Huang W, Huang J, et al. Transarterial chemoembolization combined with lenvatinib plus PD-1 inhibitor for advanced hepatocellular carcinoma: a retrospective cohort study. Front Immunol. 2022;13:848387. doi:10.3389/fimmu.2022.848387

    30. Liver Cancer Study Group of Japan; Kokudo T, Hasegawa K, Matsuyama Y, et al. Survival benefit of liver resection for hepatocellular carcinoma associated with portal vein invasion. J Hepatol. 2016;65(5):938–943. doi:10.1016/j.jhep.2016.05.044

    31. Zhang ZY, Dong KS, Zhang EL, et al. Resection might be a meaningful choice for hepatocellular carcinoma with portal vein thrombosis: a systematic review and meta-analysis. Medicine. 2019;98(50):e18362. doi:10.1097/MD.0000000000018362

    32. Kaneko S, Tsuchiya K, Yasui Y, et al. Strategy for advanced hepatocellular carcinoma based on liver function and portal vein tumor thrombosis. Hepatol Res. 2020;50(12):1375–1385. doi:10.1111/hepr.13567

    Continue Reading

  • UK economy posts surprise 0.3% growth in three months to June | Economic growth (GDP)

    UK economy posts surprise 0.3% growth in three months to June | Economic growth (GDP)

    The UK economy grew at a faster rate than expected in the second quarter, official figures show, despite a slowdown from a strong start to the year amid pressure from tax increases and Donald Trump’s global trade war.

    Figures from the Office for National Statistics showed growth in gross domestic product (GDP) slowed to 0.3% in the three months to the end of June, down from a rate of 0.7% in the first quarter.

    Although beating City forecasts of a slowdown to 0.1% growth, the latest snapshot underscores the challenge for the chancellor, Rachel Reeves, as she considers options for boosting the economy and raising revenues at her autumn budget.

    Liz McKeown, the ONS director of economic statistics, said: “Growth slowed in the second quarter after a strong start to the year. The economy was weak across April and May, with some activity having been brought forward to February and March ahead of stamp duty and tariff changes, but then recovered strongly in June.

    “Across the second quarter as a whole growth was led by services, with computer programming, health and vehicle leasing growing.

    “Construction also increased while production fell back slightly. Growth for the quarter was also boosted by updated source data for April, which while still showing a contraction, was better than initially estimated.”

    The figures were released as monthly figures showed the economy returned to growth in June, expanding by 0.4%, surpassing predictions for 0.1% growth in a recovery from two months of negative output in April and May.

    Ben Jones, the lead economist at the CBI, said: “A modest rebound in June brought Q2 to a positive close – but today’s figures confirm that the strong growth seen earlier this year was a one-off and underlying conditions remain fragile.”

    Reeves said: “Today’s economic figures are positive with a strong start to the year and continued growth in the second quarter. But there is more to do to deliver an economy that works for working people.

    “I know that the British economy has the key ingredients for success but has felt stuck for too long.

    “That is why we’re investing to rebuild our national infrastructure, cutting back on red tape to get Britain building again and boosting the national minimum wage to make work pay. There’s more to do and today’s figures only fuel my ambition to deliver on our Plan for Change.”

    Mel Stride, the shadow chancellor, said: “Any economic growth is welcome, but with business leaders saying that all indicators are flashing red, and key economists are warning that Rachel Reeves has created a £50bn black hole in the public finances, Rachel Reeves’ economic vandalism is clear.”

    skip past newsletter promotion

    Reeves has pledged to use her autumn budget to prioritise fixing Britain’s dismal productivity track record, using an exclusive article in the Guardian on Wednesday to downplay speculation over tax rises.

    While the stronger-than-expected growth performance in the second quarter will hand some relief to the chancellor, economists said it could complicate the Bank of England’s path to cutting interest rates.

    Threadneedle Street had forecast a slowdown to 0.1% in the second quarter. Last week it warned that continued growth and rising inflationary pressures driven by the surging price of food could force it to delay further cuts to borrowing costs.

    Continue Reading

  • Mongolian police seize 168 dead marmots-Xinhua

    ULAN BATOR, Aug. 14 (Xinhua) — The Mongolian police recently seized a total of 168 dead marmots, which could have spread the deadly bubonic plague, the country’s Ecological Police Department (EPD) said Thursday.

    “The marmots were found in vehicles heading to the capital city of Ulan Bator and transferred to the country’s National Center for Zoonotic Diseases for laboratory and biological tests,” the EPD said in a statement.

    While hunting marmots is illegal in Mongolia, many Mongolians ignore the law and regard the rodent as a delicacy.

    Seventeen out of all 21 Mongolian provinces are now at risk of bubonic plague, according to the National Center for Zoonotic Diseases.

    The bubonic plague is a bacterial disease that is spread by fleas living on wild rodents such as marmots, which can kill an adult in less than 24 hours if not treated in time, according to the World Health Organization.

    Continue Reading

  • HonorHealth taps longevity authority Nicholas J. Schork to lead new research division

    HonorHealth taps longevity authority Nicholas J. Schork to lead new research division

    Nicholas J. Schork, Ph.D., an international authority on human longevity and health maintenance, has joined HonorHealth Research Institute as Research Director of Longevity, Prevention and Interception.

    Dr. Schork heads a unique laboratory that is part of the Research Institute’s newly created Center for Translational Science, and his appointment is part of a significant push on the part of the Institute to expand its collaborations and the realm of precision medicine possibilities; providing specific answers to individual patients with rare or difficult to treat diseases.

    “It’s all about optimizing people’s health,” said Dr. Schork, who has trained and worked at some of the nation’s prominent health research facilities.

    Disease is not like an off and on switch; one day you don’t have disease and the next day you magically do have disease. There are processes that unfold over time that contribute to disease or reflect the pathobiology behind the disease. Research focusing on interception tries to understand those processes to the point where one can intervene on them, rather than the overt manifestations of the disease.”


    Nicholas J. Schork, PhD, Research Director of Longevity, Prevention and Interception, HonorHealth Research Institute

    How medical interception works

    For example, before treating a patient with some type of medication once they develop a specific cancer, physicians in the future might use new high-tech monitoring of a person’s genome or some type of bodily fluid such as blood, saliva or even cerebral spinal fluid to find biomarkers that would indicate early on who might be at risk of cancer.

    Such advanced interception warnings also might be attained through the use of wearable devices, imaging protocols – or anything that might provide more frequent monitoring. The physician then might recommend specific actions or medications an individual might take to avoid cancer altogether before it actually occurs and puts their overall health at risk.

    Dr. Schork said he also will employ the use of Artificial Intelligence to interrogate large electronic health records with billions of points of data to synthesize complex information and offer recommendations based on identifying patterns invisible to human clinicians.

    “Early AI evaluations show the potential to improve diagnostic accuracy, reduce errors, and personalize care pathways, provided they are validated in rigorous clinical settings,” he said, pointing to the need for clinical efforts to bolster translational discoveries, and translational science to be practiced with patients in the clinical settings.

    HonorHealth leadership enthusiastic

    In welcoming Dr. Schork and his expertise to HonorHealth, Michael Gordon, M.D., FASCO, Medical Director of the Research Institute, said: “Ultimately, it’s all about living longer, living better, reducing the risk of disease and promoting longevity. We want to emphasize prevention and interception – whether it’s intercepting in the evolution of cardiovascular disease or intercepting in the evolution of cancer – we want to find more and more sensitive ways to diagnose disease earlier; to intervene before people develop a symptomatic disorder that compromises their function; their ability to live.”

    Mark Slater, Ph.D., CEO of the Research Institute, and Vice President of Research for HonorHealth, said Dr. Schork brings with him a wealth of collaborators and facilitators from across the nation and around the world, which among other things will enhance the Institute’s partnership with Arizona State University’s new School of Medicine and Advanced Medical Engineering.

    “Nik is a brilliant mind who has really been at the forefront of developing new innovations and technologies,” Dr. Slater said. “He is a terrific data scientist, and a person in the digital and informatics space who has brought together the basic and translational science into the clinical world across a number of different diseases.”

    Sunil Sharma, M.D., MBA, Director of the Center for Translational Science, said, “I have known Dr. Schork for several years now. He is a true renaissance man and I deeply value his unique investigational abilities, scientific rigor and imagination. In his new role, working alongside me at the Center, his computational genius will be a game-changing force for innovation at our Research Institute.”

    Dr. Schork’s research background

    Most recently, Dr. Schork held the positions of Distinguished Professor and Director of the Division of Clinical Genomics and Therapeutics at Phoenix’s Translational Genomics Research Institute (TGen), a frequent clinical trial collaborator with the Institute.

    He also held the titles of: Adjunct Professor of Population Sciences as well as Molecular and Cellular Biology at City of Hope; Adjunct Professor of Psychiatry and Biostatistics at the University of California San Diego; and Adjunct Professor of Integrative Structural and Computational Biology at Scripps Research Institute.

    Prior to those positions, Dr. Schork was Professor and Director of Human Biology at the J. Craig Venter Institute; Professor, Molecular and Experimental Medicine, at Scripps Research, and Director of Bioinformatics and Biostatistics for the Scripps Translational Science Institute. Dr. Schork has also held faculty appointments at Case Western Reserve University and Harvard University.

    Dr. Schork has published over 600 articles in many areas of biomedical and translation science, including articles detailing novel methodologies and applications leveraging integrated approaches to disease diagnosis, treatment, and prevention, as well as clinical trials design. He also has a long history of collaborative and consortium-related research in which he has contributed analysis methodology and applied data analysis expertise.

    Source:

    HonorHealth Research Institute

    Continue Reading

  • Muscone suppresses inflammation and senescence of nucleus pulposus via p53 signalling during intervertebral disc degeneration

    Muscone suppresses inflammation and senescence of nucleus pulposus via p53 signalling during intervertebral disc degeneration

    Tissue sample collection

    Ethics approval, informed consent and experimental protocol was approved by the ethics committee of Shanghai Changzheng Hospital (2021SL044), and We confirm that all methods were carried out in accordance with the Declaration of Helsinki. Informed consent was provided by the patients and their relatives before harvesting intervertebral disc tissue during surgery. We also confirm that all methods relative to animal experiments were carried out in accordance with relevant guidelines and regulations. The experimental protocol was approved by the Institutional Animal Care and Use Committee of Second Military Medical University. Samples of normal intervertebral disc tissue were obtained from patients with lumbar trauma in the absence of radiological indications of degeneration, who underwent spinal fusion (Pfirrmann grade I, n = 6, age range 30 to 55 years, mean age 44 years). MRI T-2 weighted images were acquired, and the extent of intervertebral disc degeneration (IDD) was assessed utilizing the modified Pfirrmann grading system. All specimens were prepared for the isolation of nucleus pulposus cells.

    Human primary nucleus pulposus cell culture

    We performed NP cell isolation using a protocol rigorously validated by single-cell RNA sequencing (scRNA-seq)25. The collected NP tissue specimens were washed twice with sterile PBS solution. The NP region was isolated under microscopy, then minced and digested with 2 U/mL protease in DMEM medium (Gibco, NY, USA) for 20 min at 37 °C. Treating with 0.25 mg/ml type II collagenase (Gibco, Cat. No. 17101-015) for 4 h at 37 °C to obtained NP cells from the NP tissues. The remaining cell suspension was transferred into a 40 μm cell strainer (BD Biosciences, Franklin Lakes, NJ, USA) and subjected to centrifugation at 800 g for a duration of 5 min. The NP cells were subsequently resuspended in DMEM/F12 medium supplemented with 10% fetal bovine serum (FBS) (Gibco), 100 U/mL penicillin, 100 µg/mL streptomycin, and 1% L-glutamine. The cell viability exceeded 90% as determined by the cell counting kit-8 (Dojindo, Tokyo, Japan). The cells were incubated at 37 °C in an atmosphere enriched with 5% CO2, with the culture medium being refreshed every 3 days. Cells at the second passage were utilized for additional experimental methodologies.

    An in vitro model of NP cell degeneration was developed by administering recombinant human TNF-α (Peprotech, USA) (50 ng/ml) or recombinant human IL-1β (Peprotech, USA) (25 ng/ml) to the NP cells as previously reported26. For the overexpression of p53, a plasmid designed for p53 overexpression supplied by OBiO Technology (Shanghai) Corp., Ltd., or SCH529074 (MedChemExpress, USA), a p53 agonist, was utilized. Muscone (3-methylcyclopentadecanone) was purchased commercially with the purity over 98.0% (HY-N0633, MedChemExpress, China).

    CCK-8 cell viability assay

    A CCK-8 assay kit (YEASEN, Shanghai) was used to measure cell viability. Briefly, approximately 5 × 103 NP cells were seeded into each well of a 96-well plate. After treatment with the indicated group, 10 µl of CCK8 agent was added to each well according to the manufacturer’s instructions. After 1–4 h of incubation at 37 °C, the absorbances of different groups NP cells were measured with a multifunctional detection instrument (SPECTRAMAX 13X, Molecular Devices, USA).

    TUNEL staining

    Cell apoptosis was measured by transferase dUTP nick-end labeling (TUNEL) staining. Briefly, after different treatments, NP cells were fixed with 4% paraformaldehyde for 1 h and then cultured with 0.5% Triton X-100 in PBS for 10 min. After washing with PBS, the cells were incubated with reagents from a TUNEL Apoptosis Detection Kit (YEASEN, Shanghai) according to the kit’s instructions for staining. The nuclear counterstaining procedure was performed by incubating samples with 0.1 mg/mL 4’,6-diamidino-2-phenylindole (DAPI; Beyotime Biotechnology, Cat. No. C1002) in phosphate-buffered saline (PBS) for 5 min at room temperature. Apoptosis was observed under a fluorescence microscope (CKX41, Olympus, Japan), and photographs were obtained. At least five different fields in one sample was photographed to compare the effect.

    Isolation of RNA and quantitative real-time polymerase chain reaction

    RNA was isolated from human NP cells utilizing RNAiSO reagent (TAKARA, Japan) in strict adherence to the manufacturer’s protocol. Quantification of total RNA concentrations was conducted at a wavelength of 260 nm using a spectrophotometer (DU-800; Beckman Coulter, Brea, CA). Reverse transcription was performed by the PrimeScript™ RT reagent Kit with gDNA Eraser (TAKARA, Japan) in a 20-µl mixture according to the manufacturer’s instructions. TB Green® Premix Ex Taq™ (TAKARA, Japan) was used to conduct real-time PCR on a Step One Plus real-time PCR system (Applied Biosystems, Foster City, CA). GAPDH was used to normalize the mRNA expression of other genes. The comparative Ct method was used to calculate the relative expression of each transcript. Each experiment was repeated at least three times independently. The primers used in this study are listed in Supplementary Table S1.

    Western blotting

    Ice-cold lysis buffer (Cell Signaling Technology, Danvers, MA, USA) was used to harvest total protein from human NP cells. For measurement of phosphorylated protein levels, total protein was extracted at 12–24 h after treatment. A bicinchoninic acid protein assay kit (Pierce Biotechnology, Rockford, IL, USA) was used to determine the concentration of protein. Protein degeneration was prevented by using PMSF (Beyotime, China) and protease inhibitor cocktail (Meilune, China). The proteins were subjected to transfer onto PVDF membranes (Bio-Rad, CA) via electroblotting. Subsequent to this, the membranes were blocked utilizing 5% non-fat milk in TBST (50 mM Tris (pH 7.6), 150 mM NaCl, 0.1% Tween-20) and subsequently incubated in TBST containing specific antibodies overnight at 4 °C. The protein bands were visualized using SuperSignal™ West Femto Maximum Sensitivity Substrate (Thermo Fisher Scientific, USA) and Pierce™ ECL Western Blotting Substrate (Thermo Fisher Scientific, USA) to assess the expression of target proteins. The following antibodies were used: anti-MMP1 (ab52631, 1:2500 dilution), anti-MMP3 (ab53015, 1:1000 dilution), anti-MMP13 (ab39012, 1:1000 dilution), anti-ADAMTS4 (ab185722, 1:500 dilution), anti-ADAMTS5 (ab 41037, 1:250 dilution), anti-ACAN (ab3778, 1:500 dilution), anti-CHSY1 (ab153813, 1:500 dilution), anti-p53 (ab 32389, 1:10000 dilution), anti-p-p53 (ab33889, 1:1000 dilution), anti-PUMA (ab54288, 1:1000 dilution), anti-Bax (ab32503, 1:1000 dilution), anti-p21 (ab109520, 1:1000 dilution), anti-p16 (ab 108349, 1:2000 dilution), anti-CCNG2 (ab 203314, 1:500 dilution) (all from Abcam, USA) and anti-GAPDH (1:5000 dilution) (Proteintech, USA). Uncropped images were provided in supplementary Fig. 1 (Figure S1).

    High-throughput RNA sequencing and bioinformatics analysis

    Total RNA was isolated for the purpose of transcriptome sequencing utilizing the TRIzol reagent (Invitrogen, Carlsbad, USA), adhering strictly to the manufacturer’s prescribed protocol. Subsequently, the procedures encompassing library construction, RNA quality assurance, purification, quantification, and validation were executed by Shanghai NovelBio Bio-Pharm Technology Co., Ltd. For the analysis of the transcriptomic data, approximately 100 bp reads were aligned to the human genome (hg19) employing TopHat2/Bowtie2. The data were subsequently mapped to gene structures derived from RefSeq utilizing the summarize overlaps function in Intersect Strict mode (Genomic Ranges, Bioconductor). From the raw count data, reads per kilobase per million reads mapped (RPKM) values were computed for the identical gene set using Cufflinks. Differential expression analysis was conducted with the aid of edgeR, employing TMM (trimmed mean of M-values) library normalization and maintaining a false discovery rate (FDR) of 0.05. The datasets generated during the current study are available in the Gene Expression Omnibus (GEO) repository (GSE113633, Supplementary Dataset S1-3).

    Gene Ontology (GO) and KEGG pathway analyses were performed as previously described19. In summary, the p value for each Gene Ontology (GO) term was calculated employing right-sided hypergeometric tests, and the Benjamini–Hochberg method was applied for the correction of multiple tests. An adjusted p value lower than 0.05 was considered indicative of a statistically significant deviation from the anticipated distribution, suggesting enrichment of the specific GO term or pathway among the target genes. We performed GO and KEGG pathway analyses of all the differentially expressed mRNAs and GO analysis of the mRNAs that were included in the miRNA regulatory network (Supplementary Dataset S2, S3).

    Transwell invasion assay

    An invasion assay was performed with a Matrigel-coated Transwell plate to assess MMP secretion from NP cells and the migration ability of NP cells after different treatments. NP cells were plated into the upper chamber, which was coated with Matrigel (BD Biosciences, Franklin Lakes, NJ, USA). According to our previous study27the inflammatory cytokines IL-1β and TNF-α were administered 8 h post-plating at concentrations of 25 ng/ml and 50 ng/ml, respectively. After an incubation period of 48 h, cells were carefully removed from the Matrigel-coated surface of the upper chamber using swabs. The invaded cells on the lower surface of the chamber were fixed with 4% paraformaldehyde for a duration of 30 min followed by staining with 0.1% crystal violet for 10 min. Distilled deionized water was employed to wash the cells until clarity of the water was achieved. The samples were then allowed to air dry before being subjected to microscopic observation. Finally, the number of invaded cells were observed and counted using a microscope by selecting five different fields from one sample to generated the average cells.

    Flow cytometry

    Apoptotic cell measurement was conducted through flow cytometry employing an Annexin V-FITC/PI Double Staining Kit for Apoptotic Cells (BD Bioscience, USA). Cells were harvested after a 48-hour treatment period, enzymatically digested using trypsinase without EDTA, and rinsed with PBS before being processed according to the manufacturer’s protocol. Concisely, cells were resuspended in 100 µl of binding buffer, placed into a centrifuge tube, and incubated with 5 µl of dye under dark conditions for 15 min. Subsequently, 400 µl of binding buffer was added, and the samples were subjected to analysis by flow cytometry (CyAn ADP, Beckman, USA).

    Cell cycle analysis

    Cell cycle distribution was determined by flow cytometry using Cell Cycle Staining Kit (MultiSciences, Hangzhou). The NP cells were collected after 48 h of treatment, harvested by trypsinase without EDTA, washed by PBS, and followed up with the manufacturer’s instruction. Next, 1 ml DNA Staining solution and 10 µl Permeabilization solution were added to each tube, then incubate in dark for 30 min. Then the tube was detected by flow cytometry (CyAn ADP, BECKMAN, USA).

    Senescence-associated β-galactosidase (SA-β-gal) staining

    A senescence β-galactosidase staining kit (Beyotime Biotechnology, Shanghai) was used to determine the percentage of SA-β-gal-positive cells. Briefly, NP cells were cultured in 6-well plates and treated for 48 h. After fixation for 15 min, the cells were washed with PBS and then incubated with staining solution overnight. Then, the staining was observed and photographed under a fluorescence microscope (CKX31, OLYMPUS, Japan). Within each well, 500 cells were analyzed to determine the percentage of SA-β-gal-positive cells.

    Animal study

    Male C57BL/6 mice were purchased from Shanghai Model Organisms Center, Inc. (Shanghai, China). An anterior disc puncture-induced IDD animal model was established as described in our previous study27. Initially, ketamine (100 mg/kg) and acepromazine(3 mg/kg) was administered via intraperitoneal injections to induce general anesthesia, and local anesthesia was done by using bupivacaine prior to the incision procedure. The mice were systematically allocated into four distinct groups: the sham surgery group, the needle puncture group, the needle puncture combined with vehicle (1:2 PBS and 1:2 DMSO) injection group, and the needle puncture combined with muscone injection group. The ventral aspect of the intervertebral disc (IVD) was accessed through an abdominal incision. A needle was vertically introduced into the disc from the ventral side and rotated 180 degrees along its axis for a duration of 10 s. A 31-gauge needle was precisely inserted 1.5 mm parallel to the endplate, traversing the annulus fibrosus (AF) to the nucleus pulposus (NP), thereby compromising the integrity of the nucleus. In the control NC injection group, a vector of 10 µl was meticulously injected into the punctured IVD, after leaving the needle in place for five seconds, the needle was carefully removed without excessive mechanical injury. In the muscone injection group, an injection of 10ul MUSE (25 μm), with or without the addition of SCH529074 (2 μm). Magnetic resonance imaging (MRI) was conducted at intervals of 0, 3, and 9 weeks subsequent to the initial surgery. The spine of mice was analyzed in sagittal T2-weighted images using a 3.0 T clinical magnet (Philips Intera Achieva 3.0 MR). T2-weighted sections in the sagittal plane were obtained using the following settings: a fast spin echo sequence with a time to repetition (TR) of 5400 ms and a time to echo (TE) of 920 ms; a 320 (h) x 9256 (v) matrix; a field of view of 260; and 4 excitations28,29. The section thickness was 2 mm with a 0-mm gap28. The Pfirrmann grading system facilitated the evaluation of intervertebral disc (IVD) degeneration. ImageJ and Surgimap software were employed to examine the alterations in the IVD30.

    Histopathological analysis and immunohistochemical staining

    IVD sections from each group were subjected to hematoxylin and eosin (HE) staining and Safranin O-fast green (SO) staining. A histopathological grading system considering the cellularity and morphology of the AF and NP as well as the border between the two structures was used to evaluate histopathological changes31. A histopathological score of 5 indicated no degeneration, 6–10 indicated moderate degeneration and 11–15 indicated severe degeneration. Briefly, the sections were blocked with 1% (w/v) goat serum albumin at 37 °C for 1 h. Then, the sections were incubated with primary antibodies (p53, p16, p21; 1:100 dilution) overnight at 4 °C, followed by incubation with HRP-conjugated secondary antibodies for 1 h at 37 °C. The samples were imaged by a Zeiss microscope (Zeiss Axio Imager A2, Carl Zeiss Microscopy GmbH, Jena, Germany).

    Statistical analysis

    The data are presented as the mean ± standard deviation of at least three independent experiments. GraphPad Prism 6.0 software (GraphPad Software, Inc., San Diego, CA, USA) was used for statistical analysis. The normality of the data was tested using the D’Agostino-Pearson omnibus normality test. The proportion of immune-positive NP cells in human NP tissues failed to satisfy the normality test; consequently, these data were subjected to analysis using the two-tailed Mann‒Whitney U test. Conversely, the remaining datasets met the criteria for normality and were accordingly analyzed using the two-tailed Student’s t-test for the comparison between two groups, or through analysis of variance (ANOVA) followed by Tukey’s t-test for multiple group comparisons. The threshold for significance was established at 0.05.

    Continue Reading

  • Abrupt breathlessness: inebilizumab-induced Pneumocystis jirovecii pne

    Abrupt breathlessness: inebilizumab-induced Pneumocystis jirovecii pne

    Introduction

    Neuromyelitis optica spectrum disorder (NMOSD) is a rare autoimmune neurological condition which primarily manifests as recurrent optic neuritis and myelitis, with less frequent involvement of cerebral hemisphere, brainstem, and diencephalon.1,2 Repeated attacks often result in permanent neurological deficits, including blindness and paralysis.3,4 Approximately 75% of NMOSD cases harbor pathogenic autoantibodies against aquaporin-4 (AQP4), which serve as a primary effector and a diagnostic biomarker of NMOSD.5–7 The Binding of AQP4-IgG to AQP4 on astrocytes triggers classical complement activation, and antibody-dependent cellular cytotoxicity, resulting in astrocytic injury, neuronal loss, and demyelination.8,9 Additionally, B cells contribute to NMOSD pathophysiology by activating autoimmune T cells through antigen presentation, driving plasma and plasmablasts cells producing pathogenic AQP4-IgG, and secreting pro-inflammatory cytokines.10,11 Improved knowledge of the pathogenic mechanisms of NMOSD has facilitated the development of targeted therapies.

    Inebilizumab, a monoclonal antibody targeting and depleting CD19-expressing B lymphocytes, was first approved globally for use in adults with AQP4-IgG seropositive NMOSD in 2020.12 Inebilizumab has been shown excellent effectiveness in reducing the risk of attack and preventing disability progression assessed by expanded disability status scales (EDSS) in NMOSD.1 Moreover, greater reductions in NMOSD-related hospitalizations and cumulative MRI activities were observed with inebilizumab than with placebo.13,14 Beyond NMOSD, its wide B-cell depletion may benefit other neuroimmunological conditions, such as steroid-refractory neurologic immune-related adverse events and autoimmune encephalitis, which often require prolonged immunosuppression.15–17 Regarding safety, inebilizumab exhibited favorable safety in clinical trials, with most mild to moderate side effects, such as urinary tract infections, arthralgia and infusion-related reactions.18 Current evidence suggests that while anti-CD20 antibodies alone may not require routine PJP prophylaxis, the coexistence of additional risk factors (eg, high-dose corticosteroids, absolute lymphopenia, or concurrent immunosuppressants) warrants preventive measures.19 Although inebilizumab’s broader CD19 targeting theoretically poses higher infection risk than CD20-specific agents,20 no PJP cases have been reported. Here, we presented a case of an NMOSD patient that developed severe respiratory failure after inebilizumab therapy and was finally diagnosed with PJP. In addition, multiple PJP cases in patients with autoimmune disorders of the central nervous system (CNS) were summarized.

    Case Presentation

    Initial NMOSD Diagnosis

    A 41-year-old woman with NMOSD was admitted to our hospital who complained of intermittent fever and a mild cough. She denied recent travel, previous asthma, tuberculosis, tumors, and poor working or living situations. On March 8th, 2024, the patient presented skin itchiness and hyperalgesia in the right upper limb in the absence of identifiable triggers. Over time, the scratched skin developed numbness, spreading from the fingers to the right shoulder. In April, contrast-enhanced magnetic resonance imaging (MRI) of the cervical spine revealed edema in C2-C5 of the cervical spinal cord, while cranial MRI showed no abnormalities. In May, the patient experienced worsening numbness and cramps, with a new onset of numbness in her right leg. Contrast-enhanced thoracic and lumbar MRI revealed disc bulging at the L3/4, L4/5, and L5/S1 levels. The patient underwent antibody testing, confirming AQP4-IgG-seropositivity (1:32). Based on her clinical symptoms, laboratory results, and radiological features, the patient was diagnosed with NMOSD.

    B-Cell Depletion and Response

    Methylprednisolone was administered at 1000 mg for 3 days, followed by gradually tapered doses to 80 mg and maintained. Inebilizumab (300 mg) was administered on June 6th and 21st, respectively, after which the patient’s CD19+ B cells dropped from 11.04% to 0.02%, while CD20+ B cells decreased from 11.20% to 0.39% of the total lymphocytes. At the time of discharge on June 28th, her symptoms of numbness and cramps had significantly diminished with AQP4-IgG titer 1:10, and her EDSS score decreased from 2.5 to 1.5 points.

    Onset of Respiratory Symptoms and Diagnostic Workup

    However, the patient developed intermittent fever (peaking at 38.8°C) and headache since July 25th, with symptomatic fluctuations following ibuprofen administration. Upon admission on August 12th, the physical examination found an afebrile status (36.6°C) and fine moist rales in her lower lung lobes. An arterial blood gas analysis showed partial oxygen pressure (pO2) of 87 mmHg and partial pressure of carbon dioxide (pCO2) of 29 mmHg. Laboratory tests revealed a high neutrophil count (7.32*109/L) and C-reactive protein (CRP) level (37.73 mg/L), a decreased lymphocyte count at 0.66*109/L, and a low-level CD4 count of 324 cells/L. A rapid influenza virus antigen tested negative. The chest computed tomography (CT) revealed bilateral ground glass opacity with linear and patchy shadows (Figure 1). Empirical treatment with ceftriaxone and prednisone acetate (40 mg/day) was administrated. On the second day of hospitalization, the patient developed abrupt shortness of breath and her pulse oxygen saturation decreased to 87%. Due to hypoxemia (pO2 59 mmHg, pCO2 38 mmHg), she was given high-flow oxygen with a mask and atomization inhalation and was diagnosed with acute respiratory failure. To identify etiological factors, the patient underwent a series of pathogen tests (Table 1). Electric bronchoscopy was performed and bronchoalveolar lavage fluid (BALF) was collected, which was analyzed through culture, smear, galactomannan test, and metagenome next-generation sequencing (mNGS). On the third day, her axillary temperature rose to 38.7°C. Laboratory tests showed (1,3)-beta-D-glucan(G-Test) of 1378. 40 pg/mL. Additionally, mNGS revealed Pneumocystis jirovecii in BALF with reads per million (RPM) of 45776 and cytomegalovirus with 349 RPM, while no other pathogenic microorganism was detected (Table 1).

    Table 1 Pathogen Examination and Results

    Figure 1 Chest CT imaging of the patient. (A–D) On August 12, upon admission, chest CT revealed ground-glass opacities in both lungs, with linear and patchy opacities of increased density (indicated by white arrows). (a–d) Repeat chest CT on August 20 demonstrated significant resolution of the bilateral lung lesions compared to the previous scan.

    PJP Confirmation and Treatment

    Accordingly, a diagnosis of PJP was confirmed via BALF mNGS and the patient was prescribed oral trimethoprim- sulfamethoxazole (TMP-SMX) 240 mg/1200 mg every 6 hours, thymalfasin to boost immunity, and methylprednisolone (40 mg for 5 days, then decreased to 35 mg). On August 24th, the patient was free from cough and breathlessness. A repeat chest CT showed an obvious decrease in exudation (Figure 1a–d). In addition, laboratory tests revealed normal CRP and decreased G-Test level (526.50 pg/mL). She was discharged and advised for an additional four cycles of PJP prophylaxis (160 mg TMP/800 mg SMZ daily). During the two-month follow-up, the patient remained asymptomatic for respiratory symptoms. In September 2024, comparative analysis showed negative anti-AQP4 antibody, undetectable CD19 cells (0 cells/μL), and a stable EDSS score of 1.5. In October, the G-test was evaluated within normal limits.

    Discussion

    PJP Clinical Characteristics in Immunocompromised Hosts

    PJP, an opportunistic fungal infection, remains a serious threat to the immunocompromised population, especially in human immunodeficiency virus (HIV) infected individuals.21 The incidence of PJP in non-HIV-infected patients has been increasing in recent decades for the widespread use of immunosuppressive agents.22–26 Patients with PJP classically present with fever, non-productive cough and dyspnea accompanied by interstitial opacities on chest imaging.27 Nevertheless, differences exist between non-HIV and HIV-infected patients in terms of presentation, clinical progression, and outcomes. Specifically, non-HIV PJP usually exhibits more severe manifestations, evolves more rapidly, and carries a higher mortality.28–31 Early treatment for PJP is of paramount significance, for a delay of 4 days in treatment initiation has been shown to accelerate the time to death by 6.75-fold.30 Therefore, prompt and thorough examination of suspected PJP cases is crucial. To figure out the pathogen, we conducted a series of etiological tests, and eventually mNGS confirmed the diagnosis of PJP. Timely administration of anti-pneumocystis drugs prompted rapid improvement in the patient’s condition.

    B Cell Depletion and PJP Risk

    PJP is associated with low CD4+ T-cell counts, HIV infection, solid organ or stem cell transplantation, malignancies, and the use of chemotherapeutic or immunosuppressive agents.32 A decline in CD4+ T cell counts below 200 cells/μL renders highly susceptible to PJP, serving as a determinant for the initiation of prophylaxis against opportunistic infections.33–36 However, in this case, we conducted simultaneous testing of CD4+ and CD3+T cell count, ruling out PJP due to the reactive activation of T cells. Furthermore, there was no evidence of HIV infection or malignancies, and the patient had not received other immunosuppressive drugs known to cause PJP. Given the initiation of inebilizumab and corticosteroid treatment, drug-induced PJP was considered as the most probable cause of the symptoms. Based on the Naranjo Adverse Drug Reaction Scale, the probability of inebilizumab-induced PJP in the case is identified as probable (Supplementary Table 1). To the best of our knowledge, this is the first reported PJP case in an NMOSD patient following the use of inebilizumab and corticosteroids.

    Previous studies have indeed demonstrated the critical contribution of T lymphocytes response against pneumocystis, wherein T cells mediate clearance of pathogens through the recruitment and activation of effector cells.37 The increasing utilization of B cell-targeted therapies in autoimmune diseases and hematological malignancies has brought increasing attention to the role of B lymphocytes in pulmonary defense against Pneumocystis. By producing Pneumocystis-specific IgM antibodies which target fungal cell wall carbohydrates, B cells enhance pathogen opsonization, complement activation, and independent phagocytosis, thereby promoting the clearance of microbial pathogens.38 This protective role is evidenced in B-cell-deficient models, which exhibit progressively increasing Pneumocystis burden and delayed pathogen elimination.39 Clinical relevance is underscored by data from CD40 ligand knockout mice, where decreased CD19+ B cells correlated with heightened susceptibility to severe PJP, contrasting with immunocompetent mice that mount robust responses and effectively resolve infection.40 Further supporting this, clinical observations identify decreased CD19+ B-cell counts as an independent risk factor for PJP.41 Moreover, B cells contribute to post-infection immune reconstitution, facilitating bone marrow lymphocyte repopulation after pneumocystis lung infection.42 Clinically, B cell depletion with rituximab predisposes patients to both PJP and subsequent severe lung injury.43 Collectively, B cells are indispensable for effective anti-Pneumocystis, acting through antibody-dependent mechanisms and immunomodulatory pathways. Their deficiency not only impairs pathogen clearance but also disrupts immune homeostasis, exacerbating PJP severity. These insights advocate for further exploration of B cell-targeted strategies to augment host defense or modulate dysregulated inflammation in PJP.

    Corticosteroids in PJP: Dose–Response Controversies and Adjunctive Therapy Dilemmas

    Corticosteroids remain the most commonly implicated pharmacological risk factor for PJP development.44 In a retrospective analysis involving 116 HIV-negative cases, 90.5% had received corticosteroids systemically within one month before PJP diagnosis,45 corresponding with our review (Table 2). However, evidence regarding the dose of corticosteroids associated with PJP risk in non-HIV patients remains highly controversial. In a nationwide retrospective study of patients with autoimmune rheumatic diseases, the use of glucocorticoid above 10 mg daily was identified as conferring the highest PJP risk.46 Another research suggested threshold effects at more than 20 mg prednisone monotherapy, though the small sample size (n=21) may limit its generalizability.47 Moreover, a dose-dependent hierarchical pattern was observed with median glucocorticoid doses of the PJP group reaching 38.7 mg/day,48 consistent with several studies.49–51 Nevertheless, this association appears context-dependent, as low-dose prednisone (20–30 mg/day) did not significantly increase PJP risk in patients receiving concurrent rituximab.31 These inconsistencies may reflect differences in study populations and treatment regimens. In conclusion, current evidence supports a dose–response relationship, but the precise risk threshold may vary based on underlying immune status and combination therapies, underscoring the need for individualized risk assessment when considering PJP prophylaxis.

    Table 2 Overview of Pneumocystis jirovecii Pneumonia on Autoimmune CNS Disorders

    Controversy over the appropriateness of adjunctive corticosteroid therapy to PJP outcomes persists, and the optimal dosage remains inadequately explored.60 A prospective observational study including 49 intensive care units (158 cases) demonstrated that adjunctive corticosteroids increased mortality rate in non-HIV patients, but found no obvious association in HIV individuals.30 In contrast, a multicentre retrospective cohort of 139 non-HIV PJP patients indicated no significant differences between high-dose pulse methylprednisolone (500 to 1000 mg/day) and moderate-dose corticosteroid (<500 mg/day) in 30- or 180-day mortality, even in the subgroup analysis of respiratory failure cases.61 Surprisingly, a large-scale meta-analysis of 16 retrospective cohorts (2518 PJP cases) showed that low-dose corticosteroids (1 mg/kg/day prednisone equivalent) were linked to higher mortality, except in severe acute respiratory failure (PaO2<60mmHg), where corticosteroids were related to better clinical outcomes and decreased mortality.60 Conversely, others reported no significant survival benefit with adjunctive corticosteroids in moderate-to-severe PJP, irrespective of recent corticosteroid use.62 After discussion, adjunctive corticosteroid therapy was ultimately administered to the patient to prevent irreversible neurological deficits from potential subsequent attacks induced by opportunistic infections.

    PJP in CNS Autoimmune Disorders

    We conducted a comprehensive literature review of PJP patients with CNS autoimmune diseases across the PubMed database, spanning publications from its inception to 1 February 2025. The search strategy was provided in the supplementary file. A total of 12 patients (including one from our study) were included in the statistical analysis (Table 2). The median age of the patients was 32 years, ranging from 0.92 to 82 years. Despite higher prevalence among female NMOSD and multiple sclerosis (MS) patients, our retrospective analysis revealed a balanced gender distribution in PJP, potentially due to a small sample size introducing bias. The clinical presentation of PJP in this review was characterized by dyspnea (66.7%), cough (58.3%), and fever (41.7%), stressing the imperative to maintain a high index of suspicion for agnogenic respiratory symptoms in CNS autoimmune disorders. Of note, 58.3% of patients had undergone B-cell depleting therapies with PJP onset at a median of 8 weeks (range from 5 to 12 weeks) after the last treatment, indicating a potential synergistic risk between these immunosuppressive modalities possibly mediated by lymphopenia (Patients 2, 9, and 12). Diagnostically, the low sensitivity of sputum smear microscopy (8.3% in this study) for PJP correlates with typically scant sputum production. Six patients (50%) were diagnosed through BALF analysis, which demonstrates superior diagnostic efficacy of early BALF sampling for prompt diagnosis of PJP. It is necessary to acquire BALF when clinical suspicion persists despite negative sputum etiological tests. Currently, the emerging utility of mNGS further facilitates a wide detection range, particularly benefits hosts with unidentified etiologies, immunocompromised status, or infection caused by atypical or culture-negative pathogens.

    Prophylaxis Challenges and Recommendations

    Providing appropriate PJP prophylaxis treatments to immuno-compromised patients is crucial to reduce mortality and improve clinical outcomes.63 Largely based on retrospective studies involving oncology and transplant patients, a widely cited recommendation is to initiate PJP prophylaxis when receiving ≥20 mg prednisone above 2 to 4 weeks.45,64 However, the risk-benefit assessment becomes more complex in the context of CNS autoimmune diseases, such as NMOSD and MS. These patients are often managed with high-dose glucocorticoids and additional immunomodulators, yet the low incidence of PJP and non-negligible adverse effects of prophylaxis regimens have led to a lack of consensus on routine prevention.31,64 Notably, in our review of CNS autoimmune disease cases, none of the patients had received prophylaxis for PJP (Table 2), indicating that the absence of prophylactic measures may serve as a significant risk factor. Given the current gaps in evidence, large-scale prospective researches are urgently needed to evaluate the incidence of PJP in autoimmune CNS disorders, optimal thresholds, and long-term safety profiles of preventive regimens. Clinicians should weigh individual risk factors, such as cumulative immunosuppression, lymphopenia, and comorbidities when considering PJP prophylaxis in this population. A tailored approach, rather than universal prophylaxis, may offer the best balance between efficacy and safety.

    Limitations

    Our retrospective analysis has several limitations that warrant consideration. First, our review was constrained by small sample sizes and incomplete data, such as lymphocyte counts and B-cell depleting therapy duration, compromising the accuracy and generalizability of our findings. Moreover, heterogeneity in immunosuppressive regimens and baseline comorbidities among patients could introduce confounding factors, potentially obscuring real relationships between specific risk factors and PJP development. To better elucidate the risk of PJP in autoimmune CNS disorders, future research should employ prospective designs with larger patient cohorts and comprehensive documentation of immune profiles and treatment histories.

    Conclusion

    PJP is rare in autoimmune CNS disorders but may progress rapidly. Clinicians should maintain heightened vigilance for PJP and promptly obtain BALF for microbiological confirmation to facilitate early diagnosis and timely therapeutic intervention. Our report demonstrates the first confirmed NMOSD case of PJP due to maintenance therapy with inebilizumab and corticosteroids, highlighting the emerging safety concern of this highly efficacious treatment. The diagnosis of PJP in this case was unexpected, and we attributed her susceptibility to deep B cell depletion response to inebilizumab and corticosteroid therapy. Our finding supported previous research indicating the critical role of B lymphocytes in the protective immune response against PJP. The consistent temporal relationship between treatment initiation and PJP onset emphasizes the need for vigilant monitoring and proactive prophylactic strategies to mitigate this risk. Prospective multicenter studies and registries are needed to quantify risk and guide prophylaxis duration. Clinicians should consider individual risk assessment for PJP prophylaxis in NMOSD patients treated with B-cell depleting agents.

    Abbreviations

    NMOSD, neuromyelitis optica spectrum disorder; PJP, Pneumocystis jirovecii pneumonia; AQP4, aquaporin-4; EDSS, expanded disability status scales; CNS, central nervous system; MRI, magnetic resonance imaging; pO2, partial oxygen pressure; pCO2, pressure of carbon dioxide; CRP, C-reactive protein; CT, computed tomography; BALF, bronchoalveolar lavage fluid; mNGS, metagenome next-generation sequencing; G-Test, (1,3)-beta-D-glucan; TMP-SMX, trimethoprim-sulfamethoxazole; HIV, human immunodeficiency virus.

    Data Sharing Statement

    Data will be provided by the corresponding author upon reasonable request.

    Ethics Approval and Consent to Participate

    The ethics committee of Shenzhen Traditional Chinese Medicine Hospital confirmed that no separate institutional approval was required for publishing anonymized case data under local regulations.

    Consent Statement

    Written informed consent was obtained from the patient for the publication of any potentially identifiable images or data included in this case report prior to inclusion.

    Acknowledgments

    The authors thank all the clinical staff who contributed to the study.

    Funding

    This work was supported by the Sanming Project of Medicine in Shenzhen [grant numbers SZZYSM202111011]; “3030 project” of Clinical Research Program in Shenzhen Traditional Chinese Medicine Hospital in 2021 [grant numbers G3030202132].

    Disclosure

    The authors report no conflicts of interest in this work.

    References

    1. Hor JY, Asgari N, Nakashima I, et al. Epidemiology of neuromyelitis optica spectrum disorder and its prevalence and incidence worldwide. Front Neurol. 2020;11:501. doi:10.3389/fneur.2020.00501

    2. Cree B, Bennett JL, Kim HJ, et al. Inebilizumab for the treatment of neuromyelitis optica spectrum disorder (N-MOmentum): a double-blind, randomised placebo-controlled Phase 2/3 trial. Lancet. 2019;394(10206):1352–1363. doi:10.1016/S0140-6736(19)31817-3

    3. Wingerchuk DM, Banwell B, Bennett JL, et al. International consensus diagnostic criteria for neuromyelitis optica spectrum disorders. Neurology. 2015;85(2):177–189. doi:10.1212/WNL.0000000000001729

    4. Asseyer S, Schmidt F, Chien C, et al. Pain in AQP4-IgG-positive and MOG-IgG-positive neuromyelitis optica spectrum disorders. Mult Scler J Exp Transl Clin. 2018;4(3):1841750644.

    5. Lucchinetti CF, Mandler RN, McGavern D, et al. A role for humoral mechanisms in the pathogenesis of Devic’s neuromyelitis optica. Brain. 2002;125(Pt 7):1450–1461. doi:10.1093/brain/awf151

    6. Waters PJ, Pittock SJ, Bennett JL, Jarius S, Weinshenker BG, Wingerchuk DM. Evaluation of aquaporin-4 antibody assays. Clin Exp Neuroimmunol. 2014;5(3):290–303. doi:10.1111/cen3.12107

    7. Lennon VA, Kryzer TJ, Pittock SJ, Verkman AS, Hinson SR. IgG marker of optic-spinal multiple sclerosis binds to the aquaporin-4 water channel. J Exp Med. 2005;202(4):473–477. doi:10.1084/jem.20050304

    8. Nishiyama S, Misu T, Nuriya M, et al. Complement-dependent and -independent aquaporin 4-antibody-mediated cytotoxicity in human astrocytes: pathogenetic implications in neuromyelitis optica. Biochem Biophys Rep. 2016;7:45–51. doi:10.1016/j.bbrep.2016.05.012

    9. Roemer SF, Parisi JE, Lennon VA, et al. Pattern-specific loss of aquaporin-4 immunoreactivity distinguishes neuromyelitis optica from multiple sclerosis. Brain. 2007;130(Pt 5):1194–1205. doi:10.1093/brain/awl371

    10. Bennett JL, O’Connor KC, Bar-Or A, et al. B lymphocytes in neuromyelitis optica. Neurol Neuroimmunol Neuroinflamm. 2015;2(3):e104. doi:10.1212/NXI.0000000000000104

    11. Hausser-Kinzel S, Weber MS. The role of B cells and antibodies in multiple sclerosis, neuromyelitis optica, and related disorders. Front Immunol. 2019;10:201. doi:10.3389/fimmu.2019.00201

    12. Frampton JE. Inebilizumab: first approval. Drugs. 2020;80(12):1259–1264. doi:10.1007/s40265-020-01370-4

    13. Marignier R, Bennett JL, Kim HJ, et al. Disability outcomes in the N-MOMENTUM trial of inebilizumab in neuromyelitis optica spectrum disorder. Neurol Neuroimmunol Neuroinflamm. 2021;8(3). doi:10.1212/NXI.0000000000000978

    14. Cree B, Kim HJ, Weinshenker BG, et al. Safety and efficacy of inebilizumab for the treatment of neuromyelitis optica spectrum disorder: end-of-study results from the open-label period of the N-MOmentum trial. Lancet Neurol. 2024;23(6):588–602. doi:10.1016/S1474-4422(24)00077-2

    15. Rossi S, Farina A, Malvaso A, et al. Clinical course of neurologic adverse events associated with immune checkpoint inhibitors: focus on chronic toxicities. Neurol Neuroimmunol Neuroinflamm. 2024;11(6):e200314. doi:10.1212/NXI.0000000000200314

    16. Antonio M, Cerne D, Sara B, et al. Retrograde amnesia in LGI1 and CASPR2 limbic encephalitis: two case reports and a systematic literature review. Eur J Neurol. 2025;32(3):e70113. doi:10.1111/ene.70113

    17. Malvaso A, Giglio P, Diamanti L, et al. Unravelling the acute, chronic and steroid-refractory management of high-grade neurological immune-related adverse events: a call to action. Brain Sci. 2024;14(8):764. doi:10.3390/brainsci14080764

    18. Nie T, Blair HA. Inebilizumab: a review in neuromyelitis optica spectrum disorder. CNS Drugs. 2022;36(10):1133–1141. doi:10.1007/s40263-022-00949-7

    19. Zhou S, Aitken SL. Prophylaxis against pneumocystis jirovecii pneumonia in adults. JAMA. 2023;330(2):182–183. doi:10.1001/jama.2023.9844

    20. Forsthuber TG, Cimbora DM, Ratchford JN, Katz E, Stuve O. B cell-based therapies in CNS autoimmunity: differentiating CD19 and CD20 as therapeutic targets. Ther Adv Neurol Disord. 2018;11:1276983265. doi:10.1177/1756286418761697

    21. Thomas CJ, Limper AH. Pneumocystis pneumonia. N Engl J Med. 2004;350(24):2487–2498. doi:10.1056/NEJMra032588

    22. Pereira-Diaz E, Moreno-Verdejo F, De la horra C, Guerrero JA, Calderon EJ, Medrano FJ. Changing trends in the epidemiology and risk factors of pneumocystis pneumonia in Spain. Front Public Health. 2019;7:275. doi:10.3389/fpubh.2019.00275

    23. Gronseth S, Rogne T, Hannula R, Asvold BO, Afset JE, Damas JK. Epidemiological and clinical characteristics of immunocompromised patients infected with Pneumocystis jirovecii in a twelve-year retrospective study from Norway. BMC Infect Dis. 2021;21(1):659. doi:10.1186/s12879-021-06144-1

    24. Kolbrink B, Scheikholeslami-Sabzewari J, Borzikowsky C, et al. Evolving epidemiology of pneumocystis pneumonia: findings from a longitudinal population-based study and a retrospective multi-center study in Germany. Lancet Reg Health Eur. 2022;18:100400. doi:10.1016/j.lanepe.2022.100400

    25. Wu HY, Wu KS, Huang YL, et al. Identifying predictors for bacterial and fungal coinfection on chest computed tomography in patients with Pneumocystis pneumonia. J Microbiol Immunol Infect. 2021;54(4):701–709. doi:10.1016/j.jmii.2020.06.007

    26. Pates K, Periselneris J, Russell MD, Mehra V, Schelenz S, Galloway JB. Rising incidence of Pneumocystis pneumonia: a population-level descriptive ecological study in England. J Infect. 2023;86(4):385–390. doi:10.1016/j.jinf.2023.02.014

    27. Wilkin A, Feinberg J. Pneumocystis carinii pneumonia: a clinical review. Am Fam Physician. 1999;60(6):1699–1708,1713–1714.

    28. Salzer H, Schafer G, Hoenigl M, et al. Clinical, diagnostic, and treatment disparities between HIV-infected and non-HIV-infected immunocompromised patients with pneumocystis jirovecii pneumonia. Respiration. 2018;96(1):52–65. doi:10.1159/000487713

    29. Cordonnier C, Cesaro S, Maschmeyer G, et al. Pneumocystis jirovecii pneumonia: still a concern in patients with haematological malignancies and stem cell transplant recipients. J Antimicrob Chemother. 2016;71(9):2379–2385. doi:10.1093/jac/dkw155

    30. Kamel T, Janssen-Langenstein R, Quelven Q, et al. Pneumocystis pneumonia in intensive care: clinical spectrum, prophylaxis patterns, antibiotic treatment delay impact, and role of corticosteroids. A French multicentre prospective cohort study. Intensive Care Med. 2024;50(8):1228–1239. doi:10.1007/s00134-024-07489-2

    31. Park JW, Curtis JR, Jun KI, et al. Primary prophylaxis for pneumocystis jirovecii pneumonia in patients receiving rituximab. Chest. 2022;161(5):1201–1210. doi:10.1016/j.chest.2021.11.007

    32. Pike-Lee T, Syed S, Willis MA, Li Y. Pneumocystis jirovecii pneumonia in neurologic disorders: is prophylaxis necessary? Neurol Clin Pract. 2021;11(3):242–248. doi:10.1212/CPJ.0000000000000923

    33. Evernden C, Dowhan M, Dabas R, et al. High incidence of Pneumocystis jirovecii pneumonia in allogeneic hematopoietic cell transplant recipients in the modern era. Cytotherapy. 2020;22(1):27–34. doi:10.1016/j.jcyt.2019.11.002

    34. Anyimadu H, Pingili C, Sivapalan V, Hirsch-Moverman Y, Mannheimer S. The impact of absolute CD4 count and percentage discordance on Pneumocystis jirovecii pneumonia prophylaxis in HIV-infected patients. J Int Assoc Provid AIDS Care. 2018;17:1499741215. doi:10.1177/2325958218759199

    35. Phair J, Munoz A, Detels R, Kaslow R, Rinaldo C, Saah A. The risk of Pneumocystis carinii pneumonia among men infected with human immunodeficiency virus type 1. multicenter AIDS cohort study group. N Engl J Med. 1990;322(3):161–165. doi:10.1056/NEJM199001183220304

    36. Messiaen PE, Cuyx S, Dejagere T, van der Hilst JC. The role of CD4 cell count as discriminatory measure to guide chemoprophylaxis against Pneumocystis jirovecii pneumonia in human immunodeficiency virus-negative immunocompromised patients: a systematic review. Transpl Infect Dis. 2017;19(2). doi:10.1111/tid.12651

    37. Morris A, Norris KA. Colonization by Pneumocystis jirovecii and its role in disease. Clin Microbiol Rev. 2012;25(2):297–317. doi:10.1128/CMR.00013-12

    38. Rapaka RR, Ricks DM, Alcorn JF, et al. Conserved natural IgM antibodies mediate innate and adaptive immunity against the opportunistic fungus Pneumocystis murina. J Exp Med. 2010;207(13):2907–2919. doi:10.1084/jem.20100034

    39. Rong HM, Li T, Zhang C, et al. IL-10-producing B cells regulate Th1/Th17-cell immune responses in Pneumocystis pneumonia. Am J Physiol Lung Cell Mol Physiol. 2019;316(1):L291–L301. doi:10.1152/ajplung.00210.2018

    40. Hernandez-Novoa B, Bishop L, Logun C, et al. Immune responses to Pneumocystis murina are robust in healthy mice but largely absent in CD40 ligand-deficient mice. J Leukoc Biol. 2008;84(2):420–430. doi:10.1189/jlb.1207816

    41. Li XL, Liu ZX, Liu ZJ, et al. Pneumocystis pneumonia in liver transplant recipients. Am J Transl Res. 2021;13(12):13981–13992.

    42. Hoyt TR, Dobrinen E, Kochetkova I, Meissner N. B cells modulate systemic responses to Pneumocystis murina lung infection and protect on-demand hematopoiesis via T cell-independent innate mechanisms when type I interferon signaling is absent. Infect Immun. 2015;83(2):743–758. doi:10.1128/IAI.02639-14

    43. Martin-Garrido I, Carmona EM, Specks U, Limper AH. Pneumocystis pneumonia in patients treated with rituximab. Chest. 2013;144(1):258–265. doi:10.1378/chest.12-0477

    44. Ghembaza A, Vautier M, Cacoub P, Pourcher V, Saadoun D. Risk factors and prevention of Pneumocystis jirovecii pneumonia in patients with autoimmune and inflammatory diseases. Chest. 2020;158(6):2323–2332. doi:10.1016/j.chest.2020.05.558

    45. Yale SH, Limper AH. Pneumocystis carinii pneumonia in patients without acquired immunodeficiency syndrome: associated illness and prior corticosteroid therapy. Mayo Clin Proc. 1996;71(1):5–13. doi:10.4065/71.1.5

    46. Hsu HC, Chang YS, Hou TY, et al. Pneumocystis jirovecii pneumonia in autoimmune rheumatic diseases: a nationwide population-based study. Clin Rheumatol. 2021;40(9):3755–3763. doi:10.1007/s10067-021-05660-4

    47. Mecoli CA, Saylor D, Gelber AC, Christopher-Stine L. Pneumocystis jiroveci pneumonia in rheumatic disease: a 20-year single-centre experience. Clin Exp Rheumatol. 2017;35(4):671–673.

    48. Zhao Z, Huang Y, Ming B, Zhong J, Dong L. Characterization and associated risk factors of Pneumocystis jirovecii pneumonia in patients with AIRD: a retrospective study. Rheumatology. 2022;61(9):3766–3776. doi:10.1093/rheumatology/keab941

    49. Youssef J, Novosad SA, Winthrop KL. Infection risk and safety of corticosteroid use. Rheum Dis Clin North Am. 2016;42(1):157–176. doi:10.1016/j.rdc.2015.08.004

    50. Winthrop KL, Baddley JW. Pneumocystis and glucocorticoid use: to prophylax or not to prophylax (and when?); that is the question. Ann Rheum Dis. 2018;77(5):631–633. doi:10.1136/annrheumdis-2017-212588

    51. Park JW, Curtis JR, Moon J, Song YW, Kim S, Lee EB. Prophylactic effect of trimethoprim-sulfamethoxazole for pneumocystis pneumonia in patients with rheumatic diseases exposed to prolonged high-dose glucocorticoids. Ann Rheum Dis. 2018;77(5):644–649. doi:10.1136/annrheumdis-2017-211796

    52. Garcia-Moreno J, Igartua LJ, Montes RM. Pneumocystis Jirovecii pneumonia in a patient with anti-N-methyl-D-aspartate receptor Postherpetic Encephalitis. Pediatr Infect Dis J. 2016;35(7):816–817. doi:10.1097/INF.0000000000001165

    53. Yann K, Jackson F, Sharaf N, et al. Acute respiratory distress syndrome following alemtuzumab therapy for relapsing multiple sclerosis. Mult Scler Relat Disord. 2017;14:1–3. doi:10.1016/j.msard.2017.03.001

    54. Lau AY, Lui G, Chan KP, Au C, Mok V, Ziemssen T. Pneumocystis pneumonia in a patient treated with alemtuzumab for relapsing multiple sclerosis. Mult Scler Relat Disord. 2020;38:101503. doi:10.1016/j.msard.2019.101503

    55. Sadeghi HZM, Mehdinezhad H, Mehraeen R, Golparvar AM, Tavakoli PA. Pneumocystis jirovecii pneumonia complicated a case of SARS-CoV-2 infection and multiple sclerosis after treatment with rituximab. Clin Case Rep. 2023;11(6):e7455. doi:10.1002/ccr3.7455

    56. Nakano H, Motoyama R, Tanaka K, Tanaka M. A case of neuromyelitis optica with varicella zoster virus meningitis during mitoxantrone treatment. Rinsho Shinkeigaku. 2011;51(9):703–705. doi:10.5692/clinicalneurol.51.703

    57. Zhou J, Zhang Y, Jin Y, et al. Pneumocystis pneumonia during Rituximab treatment in children with autoimmune nerve system diseases. J Neuroimmune Pharmacol. 2018;13(3):277–278. doi:10.1007/s11481-018-9791-8

    58. Liu L, Huang Y, Shu S, Zhou H, Fang F, Liu X. Nocardia farcinica pneumonia complicated by pneumocystis jiroveci infection in children with Neuromyelitis optica spectrum disorders: a case report and literature review. Ital J Pediatr. 2024;50(1):255. doi:10.1186/s13052-024-01827-2

    59. Baulier G, Issa N, Gabriel F, Accoceberry I, Camou F, Duffau P. Guidelines for prophylaxis of Pneumocystis pneumonia cannot rely solely on CD4-cell count in autoimmune and inflammatory diseases. Clin Exp Rheumatol. 2018;36(3):490–493.

    60. Ding L, Huang H, Wang H, He H. Adjunctive corticosteroids may be associated with better outcome for non-HIV Pneumocystis pneumonia with respiratory failure: a systemic review and meta-analysis of observational studies. Ann Intensive Care. 2020;10(1):34. doi:10.1186/s13613-020-00649-9

    61. Morimoto Y, Matsui H, Fujioka H, et al. Effectiveness of pulse methylprednisolone in patients with non-human immunodeficiency virus pneumocystis pneumonia: a multicentre, retrospective registry-based cohort study. BMC Infect Dis. 2024;24(1):1233. doi:10.1186/s12879-024-10151-3

    62. Moon SM, Kim T, Sung H, et al. Outcomes of moderate-to-severe Pneumocystis pneumonia treated with adjunctive steroid in non-HIV-infected patients. Antimicrob Agents Chemother. 2011;55(10):4613–4618. doi:10.1128/AAC.00669-11

    63. Gaborit BJ, Tessoulin B, Lavergne RA, et al. Outcome and prognostic factors of Pneumocystis jirovecii pneumonia in immunocompromised adults: a prospective observational study. Ann Intensive Care. 2019;9(1):131. doi:10.1186/s13613-019-0604-x

    64. Schmajuk G, Jafri K, Evans M, et al. Pneumocystis jirovecii pneumonia (PJP) prophylaxis patterns among patients with rheumatic diseases receiving high-risk immunosuppressant drugs. Semin Arthritis Rheum. 2019;48(6):1087–1092. doi:10.1016/j.semarthrit.2018.10.018

    Continue Reading

  • Hapag-Lloyd posts 3.1% lower net profit in H1, lowers top of FY forecast – Reuters

    1. Hapag-Lloyd posts 3.1% lower net profit in H1, lowers top of FY forecast  Reuters
    2. Hapag-Lloyd: Good growth and solid half-year results in a volatile market  Hapag-Lloyd
    3. Hapag-Lloyd posts 3.1% lower net profit in H1, lowers top of FY forecast By Reuters  Investing.com
    4. Hapag-Lloyd (ETR:HLAG) jumps 3.9% this week, though earnings growth is still tracking behind five-year shareholder returns  Yahoo Finance
    5. Hapag-Lloyd Narrows Guidance Despite Heightened Uncertainty From Global Tensions  The Wall Street Journal

    Continue Reading

  • Kate Moss to present BBC Radio 6 Music David Bowie podcast series | Media

    Kate Moss to present BBC Radio 6 Music David Bowie podcast series | Media

    BBC Radio 6 Music – the UK’s No.1 digital station – has revealed a slate of new programmes for autumn 2025.

    Kate Moss will host Music Uncovered, David Bowie: Changeling, a world exclusive, eight-part podcast exploring David Bowie’s artistic evolution between 1970-1975.

    Kate Moss said: “David Bowie was a very special person. Someone who was much more than a friend – he was an enigma. So, when the chance came to dive into this extraordinary five-year chapter of Bowie’s life for 6 Music and BBC Sounds, hearing from those who joined him on his creative journey and those he continues to inspire, I was excited to help share the story of such an incredible transformation. This podcast is a real celebration of my friend, a true British icon.”

    Will Wilkin, commissioning executive, BBC Music for David Bowie: Changeling, said: “David Bowie and Kate Moss both transcend fashion, music, and art – true creative forces constantly reshaping culture. This podcast offers a personal look at Bowie’s transformation, revealing his impact and legacy nearly a decade on. 

    “With Kate’s narration, the Bowie archive at V&A East Storehouse, 6 Music’s connection to his sound, and Zinc Media’s award-winning production, David Bowie: Changeling gathers the voices of those he inspired. As Bowie said, ‘I don’t know where I’m going from here, but I promise it won’t be boring’ – this is Kate Moss on that constant reinvention that made him an icon.”

    Available on BBC Sounds from 6am on September 10 and broadcast on 6 Music on September 22 (12-2am), the podcast features rare and unheard archive interviews with Bowie, including an exclusive interview from 2001 with podcast creator Des Shaw, and recently unearthed audio from the BBC Archive. 

    It also includes new interviews with those who knew Bowie, and who continue to be inspired by his legacy, including Boy George, Chrissie Hynde, Dave Gahan, Edward Enninful, Elton John, Goldie, Iggy Pop, Harris Reed, Robbie Williams and Twiggy, with archive interviews from Lady Gaga, Sinéad O’Connor, Lou Reed, Tracey Emin and more. 

    Episodes explore Bowie’s early performances with The Hype; his fascination with Andy Warhol; the creation of The Rise And Fall Of Ziggy Stardust and The Spiders From Mars and the legacy of his iconic Ziggy Stardust image; the famous Hammersmith Odeon show where he retired the rock persona; The 1980 Floor Show at London’s Marquee Club in 1973; the recording of Diamond Dogs; and his creation of The Thin White Duke.

    The series comes as BBC partners, the V&A, launch the David Bowie Centre at V&A East Storehouse. The David Bowie Centre is a new working archive for the world’s largest collection dedicated to Bowie’s life and works, with free rotating displays of highlights from the collection. 

    David Bowie and Kate Moss both transcend fashion, music, and art – true creative forces constantly reshaping culture

    Will Wilkin

    In a further programming announcement, Beth Ditto has joined 6 Music with a new series, Indie Forever Disco, and a regular spot on Nick Grimshaw’s Breakfast Show titled What Would Beth Ditto Do? Grimshaw reflected on a less-than-successful pairing with Ditto on 6 Music during Glastonbury in our Music Week Awards interview following the station’s victory.

    Beth Ditto will join Nick Grimshaw on the 6 Music Breakfast Show (weekdays 7-10am) every Tuesday from September 16. The singer, songwriter and Gossip frontwoman will share her take on life’s dilemmas and respond to listeners’ quandaries. What Would Beth Ditto Do? will also be available in vision each week on 6 Music’s YouTube channel.

    Beth Ditto said: “I’m so excited to be joining 6 Music and to get to work with Nick every week is just the cherry on top. I’ve always been a huge fan of the station, so honestly… I’m so excited, I’m speechless. Which should work out great for radio!”

    In November, Ditto will also host a new series, Indie Forever Disco with Beth Ditto for BBC Sounds, in which she’ll celebrate the biggest and best indie hits for the dance floor – from classic tracks to the hits of tomorrow.

    6 Music has also unveiled a 12-part series titled Grounding, in which four neurodivergent artists share their lived experiences: composer, DJ and 6 Music’s Afrodeutsche, author and podcaster Blindboyboatclub, musician, producer and DJ Emma-Jean Thackray and singer, songwriter and musician Gary Numan.

    Grounding will be broadcast Monday to Thursday, 11pm-1am from September 15 to October 2.  Emma-Jean Thackray (September 15-17), Gary Numan (September 18 and 22-23), Blindboyboatclub (September 24, 25 and 29) and Afrodeutsche (September 30 and October 1-2) each present three two-hour episodes.

    Across the series, they share their stories, explore how their neurodivergence shapes their relationship with music and highlight the songs that hold personal significance.

    Grounding is produced by Granny Eats Wolf, working with The Donaldson Trust, The national body for neurodiversity, which delivers services and improves access and opportunity for neurodivergent people.

    Samantha Moy, head of BBC Radio 6 Music, said: “There’s so much to look forward to on 6 Music this autumn. It’s a joy to welcome Kate Moss to celebrate an artist close to her heart and ours – David Bowie. I’m proud to be working with four incredible neurodivergent artists, Emma-Jean Thackray, Gary Numan, Blindboyboatclub and our very own Afrodeutsche, who will lead conversations exploring music, identity and creativity in our new series, Grounding. And then there’s Beth Ditto, who will be doing all of that and more with Nick Grimshaw on our Breakfast Show, and later this year with Indie Forever Disco.”

    PHOTO: Sukita

     

    Continue Reading

  • Decisive phase of conflict with India brought honour to Pakistan: Dar

    Decisive phase of conflict with India brought honour to Pakistan: Dar

    LAHORE  –  Deputy Prime Minister and Foreign Minister Senator Mohammad Ishaq Dar said on Wednesday that Pakistan is making rapid progress and, by the grace of Allah Almighty, this journey will continue until the country emerges as a strong fortress of Islam on the world map. He expressed confidence that with the prayers of people, Pakistan will achieve greater strength, prosperity, and dignity in the international community.

    Talking to the media after inaugurating the 982nd Urs of Hazrat Ali Hajveri, known as Data Ganj Bakhsh, Ishaq Dar extended greetings on behalf of PML-N Quaid Mian Nawaz Sharif, President Asif Ali Zardari, Prime Minister Shehbaz Sharif, and Chief Minister Punjab Maryam Nawaz Sharif. He added that this year’s Urs coincides with Pakistan’s Independence Day, making the occasion especially meaningful. He urged devotees to offer special prayers, particularly during the concluding session late Friday night, for Pakistan’s growth, prosperity, and dignity.

    The deputy prime minister acknowledged that development is a long process but stressed that Pakistan’s progress has already begun. He expressed gratitude to Allah Almighty for bestowing honour upon Pakistan and guiding it toward economic stability and diplomatic success. Quoting the Holy Prophet (PBUH), he reminded participants to pray with full faith that their prayers will be accepted. To a question, Ishaq Dar said that among the many blessings Allah Almighty has granted Pakistan, the greatest was the victory in Maarka-e-Haq.

    He said the decisive phase of the conflict, fought from May 6 night to May 10 morning on land, at sea, and in the air, brought immense honour to the country. “The world now looks toward Pakistan with respect and admiration,” he remarked.

    To another query, he said that others should answer for their own actions, adding that Pakistan’s focus must remain on effectively managing its affairs and moving forward. He described Pakistan as a peaceful country with a peace-loving government and said Prime Minister Shehbaz Sharif is fully focused on economic development. Under the Prime Minister’s leadership and guidance, his team is working diligently to advance quickly on economic, diplomatic, and developmental fronts.

    On counterterrorism, he revealed that on Tuesday, the Ministry of Foreign Affairs hosted an extensive dialogue with a high-level US delegation comprising about two dozen officials from both countries. He said Pakistan had been providing Washington with irrefutable evidence for the past year that the Majeed Brigade is part of the BLA and was involved in the Jaffar Express attack. “We had shared so much credible evidence that the US was left with no option but to designate them as a Foreign Terrorist Organisation,” he said.

    On the issue of water, he reaffirmed Pakistan’s long-standing position, first clearly articulated on April 24 at the National Security Committee meeting chaired by Prime Minister Shehbaz Sharif, that any attempt to block or divert Pakistan’s water will be considered an act of war. “Pakistan’s 240 million people cannot afford to lose even a single drop of the water that is rightfully theirs,” he said, adding that the country will use all necessary diplomatic means to safeguard its water rights.

    Earlier, the foreign minister officially inaugurated the Urs celebrations by laying the traditional chador at the saint’s shrine and offering Fateha. The three-day Urs, from Wednesday to Friday, is drawing thousands of devotees from across Punjab and other provinces.

    Federal Minister for Communications and Istehkam-e-Pakistan Party (IPP) President Abdul Aleem Khan, Punjab Minister for Food, Housing and Urban Development Bilal Yasin, PML-N leader Mian Marghoob Ahmad, and Secretary Auqaf Dr. Tahir Raza Bukhari also attended.

    The ministers welcomed Ishaq Dar on his arrival at Data Darbar and joined the opening prayers. Comprehensive security arrangements have been implemented to ensure the smooth conduct of the Urs.


    Continue Reading

  • Prenatal acetaminophen linked to higher risk of autism and ADHD

    Prenatal acetaminophen linked to higher risk of autism and ADHD

    Researchers at the Icahn School of Medicine at Mount Sinai have found that prenatal exposure to acetaminophen may increase the risk of neurodevelopmental disorders, including autism spectrum disorder and attention-deficit/hyperactivity disorder (ADHD), in children.

    The study, published today in BMC Environmental Health, is the first to apply the rigorous Navigation Guide methodology to systematically evaluate the rigor and quality of the scientific literature.

    Acetaminophen (often sold under the brand name Tylenol®, and known as paracetamol outside the United States and Canada) is the most commonly used over-the-counter pain and fever medication during pregnancy and is used by more than half of pregnant women worldwide.

    Until now, acetaminophen has been considered the safest option for managing headache, fever, and other pain. Analysis by the Mount Sinai-led team of 46 studies incorporating data from more than 100,000 participants across multiple countries challenges this perception and underscores the need for both caution and further study.

    The Navigation Guide Systematic Review methodology is a gold-standard framework for synthesizing and evaluating environmental health data. This approach allows researchers to assess and rate each study’s risk of bias, such as selective reporting of the outcomes or incomplete data, as well as the strength of the evidence and the quality of the studies individually and collectively.

    Our findings show that higher-quality studies are more likely to show a link between prenatal acetaminophen exposure and increased risks of autism and ADHD. Given the widespread use of this medication, even a small increase in risk could have major public health implications.”


    Diddier Prada, MD, PhD, Assistant Professor, Population Health Science and Policy, and Environmental Medicine and Climate Science, Icahn School of Medicine, The Mount Sinai Hospital

    The paper also explores biological mechanisms that could explain the association between acetaminophen use and these disorders. Acetaminophen is known to cross the placental barrier and may trigger oxidative stress, disrupt hormones, and cause epigenetic changes that interfere with fetal brain development.

    While the study does not show that acetaminophen directly causes neurodevelopmental disorders, the research team’s findings strengthen the evidence for a connection and raise concerns about current clinical practices.

    The researchers call for cautious, time-limited use of acetaminophen during pregnancy under medical supervision; updated clinical guidelines to better balance the benefits and risks; and further research to confirm these findings and identify safer alternatives for managing pain and fever in expectant mothers.

    “Pregnant women should not stop taking medication without consulting their doctors,” Dr. Prada emphasized. “Untreated pain or fever can also harm the baby. Our study highlights the importance of discussing the safest approach with health care providers and considering non-drug options whenever possible.”

    With diagnoses of autism and ADHD increasing worldwide, these findings have significant implications for public health policy, clinical guidelines, and patient education. The study also highlights the urgent need for pharmaceutical innovation to provide safer alternatives for pregnant women.

    Source:

    Journal reference:

    Prada, D., et al. (2025) Evaluation of the evidence on acetaminophen use and neurodevelopmental disorders using the Navigation Guide methodology. Environmental Health. doi.org/10.1186/s12940-025-01208-0

    Continue Reading