Leo Pharma mengambil alih perkembangan global spevigo untuk penyakit kulit langka GPP

Thornhill, Ontario, Kanada – 31 Oktober 2020: Leo Pharma Kanada menandatangani di luar gedung kantor pusat mereka di Thornhill, Ontario, Kanada. Leo Pharma seperti perusahaan farmasi Denmark multinasional | Kredit Gambar: © Jhvephoto – Stock.adobe.com.

Leo Pharma telah menandatangani perjanjian lisensi dan transfer global dengan Boehringer Ingelheim untuk komersialisasi dan pengembangan spesolimab yang berkelanjutan, dipasarkan sebagai Spevigo (1). Antibodi monoklonal (MAb) adalah perlakuan target pertama yang disetujui untuk suar psoriasis pustular (GPP) umum, kondisi kulit inflamasi yang langka dan berpotensi mengancam jiwa.

Perjanjian tersebut menempatkan Leo Pharma di pucuk pimpinan fase Spevigo berikutnya, membangun kehadirannya di lebih dari 40 negara – termasuk Amerika Serikat, Jepang, Cina, dan di seluruh Eropa – di mana ia disetujui untuk pengobatan suar GPP pada orang dewasa. Otorisasi peraturan juga telah diberikan untuk indikasi GPP yang lebih luas di AS, Uni Eropa, dan Cina (2).

Menargetkan kebutuhan yang tidak terpenuhi pada penyakit dermatologis langka

Kunci takeaways

· Leo Pharma akan memimpin komersialisasi global dan pengembangan Spevigo, memperluas perannya dalam manufaktur dan akses obat dermatologi langka.

· Antibodi monoklonal penargetan IL-36 adalah maju melampaui GPP, dengan upaya pengembangan baru yang difokuskan pada penyakit kulit autoinflamasi langka lainnya.

· Perjanjian tersebut meliputi tonggak sejarah dan royalti, menyoroti tren pembuatan kesepakatan strategis di farmasi untuk terapi penyakit langka dengan kebutuhan yang tidak terpenuhi tinggi.

Spesolimab adalah mAb yang dimanusiakan dan selektif yang menghalangi aktivasi reseptor interleukin-36 (IL-36), jalur pensinyalan pusat untuk pengembangan beberapa penyakit kulit autoinflamasi (3-5). Di GPP, jalur IL-36 dianggap sebagai pendorong kritis aktivitas penyakit, menjadikannya target utama untuk intervensi terapeutik.

GPP adalah kondisi yang parah dan tidak dapat diprediksi yang ditandai oleh pustula steril yang meluas, peradangan kulit, dan gejala sistemik seperti demam, kelelahan, dan dalam beberapa kasus, gagal organ (6-10). Pasien mungkin mengalami kambuh atau penyakit persisten dengan intensitas yang bervariasi, dan kualitas hidup seringkali mengalami gangguan signifikan. Studi klinis menunjukkan GPP berbeda dari psoriasis plak, dengan patologi dan kebutuhan pengobatan yang unik (6,7).

Spevigo telah dievaluasi dalam program pengembangan klinis terbesar yang didedikasikan untuk GPP hingga saat ini (1). Itu tetap satu-satunya pengobatan untuk suar GPP yang secara khusus menargetkan IL-36 dan telah divalidasi dalam uji coba acak yang dikendalikan plasebo. Investigasi lebih lanjut sedang dilakukan untuk menilai potensinya pada penyakit dermatologis yang dimediasi IL-36 lainnya dengan opsi pengobatan terbatas.

Pergeseran strategis untuk memperluas akses global

Berdasarkan perjanjian tersebut, Leo Pharma memikul tanggung jawab untuk komersialisasi global dan pengembangan klinis lebih lanjut dari obat (1). Perusahaan berencana untuk memanfaatkan infrastruktur dermatologi medis yang ada untuk meningkatkan kesadaran dan akses pasien ke perawatan.

“Bermitra untuk membawa Spevigo ke lebih banyak pasien lebih dari sekadar langkah strategis – itu berarti kesempatan untuk membantu orang yang hidup dengan GPP dengan mengatasi penyakit dengan pilihan perawatan terbatas dan bertujuan untuk meningkatkan kualitas hidup mereka,” kata Christophe Bourdon, CEO Leo Pharma (1). “Kami terinspirasi oleh upaya perintis Boehringer Ingelheim dan senang membangun di atas fondasi itu.”

Kesepakatan itu diperkirakan akan ditutup pada paruh kedua tahun 2025, clearance regulasi yang tertunda (1). Boehringer Ingelheim akan menerima pembayaran di muka sebesar EUR 90 juta, selain pembayaran tonggak potensial dan royalti berjenjang berdasarkan kinerja komersial.

Pekerjaan dasar dan arah masa depan

Perkembangan awal Spevigo Boehringer Ingelheim menandai tonggak penting bagi pasien GPP dan dermatologi medis (1).

“Kami sangat bangga dengan apa yang diwakili Spevigo bagi orang -orang yang hidup dengan GPP dan membawa inovasi kepada komunitas pasien ini telah menjadi cerminan yang kuat dari tujuan kami untuk mengubah kehidupan,” kata Shashank Deshpande, ketua dewan direksi pengelola dan kepala farmasi manusia di Boehringer Ingelheim (1). “Spevigo memegang janji yang signifikan, dan memastikannya mencapai potensi penuhnya membutuhkan fokus yang berkelanjutan, dan keahlian dalam dermatologi medis. Dengan lebih dari enam dekade dedikasi tunggal untuk bidang ini, Leo Pharma sangat diposisikan dengan baik untuk membangun di atas fondasi yang kuat yang telah kami letakkan.”

GPP tetap merupakan kondisi yang sangat kompleks dan kurang terlayani (1). Tingkat kematian untuk kasus yang parah dapat berkisar dari 2% hingga 16%, seringkali karena komplikasi seperti kegagalan organ multisistem dan sepsis. Banyak pasien juga mengalami komorbiditas yang meningkatkan beban sistem pribadi dan perawatan kesehatan.

Referensi

  1. Leo Pharma. Boehringer Ingelheim dan Leo Pharma memasuki kemitraan untuk mengkomersialkan dan lebih lanjut mengembangkan Spevigo (Spesolimab). Siaran pers. 14 Juli 2025.
  2. Boehringer Ingelheim. Komisi Eropa menyetujui Spevigo untuk indikasi baru dan diperluas dalam psoriasis pustular umum. Siaran pers. 30 September 2024.
  3. Morita. A.; Strober, B.; Beban, iklan; et al. Kemanjuran dan keamanan spesolimab subkutan untuk pencegahan general psoriasis flare (Effisayil 2): uji coba internasional, multisenter, acak, terkontrol plasebo. Lanset. 2023 402: 1541–1551.
  4. Choon, S.; Lebwohl, M.; Marrakchi, S.; et al. Protokol studi Global Effisayil 1 Fase II, multisenter, acak, double-blind, percobaan spesolimab terkontrol plasebo pada pasien dengan psoriasis pustular umum yang disajikan dengan suar akut. Bmj terbuka. 2021 11: E043666.
  5. Bachelez, H.; Choon, S.; Marrakchi, S.; et al. Percobaan spesolimab untuk psoriasis pustular umum. N engl j with. 2021 385: 2431–2440.
  6. Marrakchi, S.; Puig, L. Patofisiologi psoriasis pustular umum. Am J Clin Dermatol. 2022 23: 13–19.
  7. Prinz, J.; Choon, S.; Griffiths, C.; et al. Prevalensi, komorbiditas dan kematian psoriasis pustular umum: tinjauan literatur. J eur acad dermatol venereol. 2023 37: 256–273.
  8. Choon, S.; Navarini, A.; Pinter, A. Kursus klinis dan karakteristik psoriasis pustular umum. Am J Clin Dermatol. 2022 23: 21–29.
  9. Gooderham, M.; Van Voorhees, A.; Lebwohl, M. Pembaruan tentang psoriasis pustular umum. Expert Rev Clin Immunol. 2019 15: 907–919.
  10. Reisner, D.; Johnsson, F.; Kotowsky, N.; et al. Dampak psoriasis pustular umum dari perspektif orang yang hidup dengan kondisi: hasil survei online. Am J Clin Dermatol. 2022 23: 65–71.

Kolaborasi generatif yang digerakkan AI untuk perawatan penelitian untuk komplikasi perdarahan otak

Cedera otak, darah pada otak yang terluka (stroke, perdarahan otak, perdarahan intraserebral, perdarahan subaraknoid, malformasi arteriovenous, ruptur aneurisma otak, cedera otak traumatis) | Kredit Gambar: © A banyak pica – stock.adobe.com

Proteros biostructures GmbH, an early-stage drug discovery services provider in Germany, has announced an agreement with Qanatpharma of Switzerland, Zuse Institute Berlin (ZIB), and Ukraine-based Enamine to launch a research collaboration intending to leverage innovations from all four partners—notably, ZIB's artificial intelligence (AI)-based generative ligand design—to accelerate discovery of novel Terapi yang menargetkan defisit perfusi serebral yang terkait dengan perdarahan subarachnoid (SAH) (1).

Mitra mengatakan dalam siaran pers pada 9 Juli 2025 bahwa mereka berharap kolaborasi akan menetapkan preseden untuk AI generatif untuk digunakan dalam penemuan tahap awal untuk mengobati kondisi serebrovaskular (1). Fokus awal kemitraan adalah target protein yang diidentifikasi oleh qanatpharma yang membantu mengatur resistensi serebrovaskular di otak, mekanisme yang telah terbukti terganggu pada pasien dengan SAH, yang merupakan bentuk stroke yang parah.

Kebutuhan yang tidak terpenuhi untuk penderita stroke

Kunci takeaways

  • Proteros, Qanatpharma, Zuse Institute Berlin, dan Enamine telah bermitra untuk menerapkan AI generatif untuk penemuan obat tahap awal untuk mengobati masalah perfusi otak dalam perdarahan subarachnoid (SAH).
  • Target kolaborasi menunda iskemia serebral, komplikasi SAH yang umum dan saat ini tidak tertangani, yang bertujuan untuk meningkatkan hasil.
  • Skrining majemuk sedang berlangsung, dengan in-vitro Validasi yang diharapkan kemudian pada tahun 2025, memperkuat tren industri yang lebih luas untuk mengintegrasikan alat AI generatif secara cermat.

Komplikasi terkemuka, dan sebelumnya kebutuhan yang tidak terpenuhi, karena SAH yang selamat ditunda iskemia serebral (DCI), yang mengurangi perfusi otak dan dapat mengakibatkan kerusakan neurologis jangka panjang, kecacatan, atau kematian (1). Standar perawatan saat ini untuk SAH gagal menargetkan mekanisme molekuler yang mendorong terjadinya DCI.

“Tertunda iskemia serebral adalah kontributor utama untuk hasil yang buruk setelah perdarahan subarachnoid, dan pilihan pengobatan saat ini tetap terbatas,” Steffen-Sebastian Bolz, MD, PhD, kepala petugas medis dan ilmiah di Qanatpharma, mengatakan dalam rilis (1). “Dengan membangun konsorsium ini, kami menyatukan insinyur pembelajaran mesin yang paling cerdas, ahli kimia, ahli biologi struktural, dan ilmuwan lain untuk mengatasi komplikasi kritis ini. Bersama-sama, kami bertujuan untuk mempercepat pengembangan terapi yang ditargetkan yang dapat secara signifikan meningkatkan pemulihan dan hasil jangka panjang untuk pasien yang terkena SAH.”

Keempat mitra mengatakan mereka sudah memulai upaya penyaringan majemuk dan berencana untuk melanjutkan in-vitro Studi validasi sebagai paruh kedua dari 2025 berlangsung (1).

Perusahaan yang berkolaborasi dalam inovasi AI

Other partnerships involving, at least in part, the adoption of generative-AI approaches have been announced in 2025. In February, two United States companies, Delaware-based Incyte and California-based Genesis Therapeutics, agreed to a strategic collaboration making use of generative and predictive AI to help research, discover, and develop novel small-molecule medicines for various targets (2).

Beberapa perusahaan sedang mengerjakan teknologi generatif-AI ke dalam alur kerja mereka bahkan ketika mereka tetap berhati-hati tentang penggunaan berlebihan atau paparan berlebih. Pada bulan Mei 2025, Emerson yang berbasis di St. Louis, yang sedang dalam akuisisi Teknologi Aspen, mengumumkan pendekatan baru yang digerakkan AI yang bertujuan meningkatkan keandalan dan kinerja operasi manufaktur kritis misi, termasuk yang ada di sektor farmasi (3). Namun, pada saat yang sama, perusahaan memperingatkan bahwa alat-alat generatif-AI publik-setidaknya sampai pada titik ini, setidaknya-tidak cocok untuk pengaturan industri karena masalah keamanan dan keandalan.

Sebagai bagian dari Teknologi FarmasiSeri “Outlook Industri” ® Group untuk tahun 2025, Preeya Beczek, direktur pelaksana dan salah satu pendiri Beczek.com Ltd, seorang spesialis dalam urusan dan kepatuhan peraturan, secara keseluruhan, AI generatif adalah tren teratas untuk ditonton karena berkaitan dengan cara-cara di mana perusahaan dalam operasi industri (4).

“Saya pikir kita hanya akan memiliki lebih banyak orang mengadopsi (AI generatif dan) cara kerja, dalam pekerjaan sehari-hari mereka,” kata Beczek dalam wawancara (4). “Saya pikir akan ada beberapa dorongan untuk mendapatkan kecepatan, (dalam) hal -hal seperti pembuatan jadwal, (jadi) mereka menjadi lebih pendek.”

Klik di sini untuk melihat wawancara lengkap.

Referensi

1. Proteros Biostruktur GmbH. Qanatpharma, Zuse Institute Berlin, Enamine, dan Proteros Biostruktur mengumumkan kolaborasi penemuan utama yang digerakkan oleh generatif. Siaran pers. 9 Juli 2025.
2. Incyte. Incyte dan Genesis Therapeutics mengumumkan kolaborasi penelitian yang berfokus pada AI strategis. Siaran pers. 20 Februari 2025.
3. Emerson. Portofolio AI yang diperluas Emerson membuka jalan bagi operasi otonom yang lebih dioptimalkan. Siaran pers. 22 Mei 2025.
4. Thomas, F. Outlook Industri 2025: Personalisasi Kedokteran Presisi dan Mengadopsi AI. Pharmtech.com20 Februari 2025.

FDA menerbitkan lebih dari 200 surat tanggapan lengkap

Silver Spring, MD, USA – 25 Juni 2022: Kampus Oak White FDA, markas besar Administrasi Makanan dan Obat -obatan Amerika Serikat, sebuah agen federal dari Departemen Kesehatan dan Layanan Kemanusiaan (HHS) | Kredit Gambar: © TADA Images – stock.adobe.com

Pada 10 Juli 2025, FDA membuat publik lebih dari 200 surat keputusan rahasia yang sebelumnya – dikenal sebagai Lengkapi Surat Respons Lengkap (CRLS) – dikeluarkan antara tahun 2020 dan 2024 untuk aplikasi obat dan biologis yang tidak disetujui selama siklus peninjauan awal mereka (1). Surat -surat ini memberikan alasan terperinci mengapa suatu aplikasi tidak dapat disetujui dalam bentuk yang diajukan, termasuk masalah yang terkait dengan keamanan, kemanjuran, kekurangan manufaktur, dan bioequivalensi (1).

Langkah yang belum pernah terjadi sebelumnya

Sebelumnya, FDA tidak secara rutin berbagi CRL di depan umum. Sponsor sering mengungkapkan informasi minimal saat mengumumkan tidak persetujuan. Sebuah studi cross-sectional 2015 yang diterbitkan di BMJ menemukan bahwa pengembang obat menghilangkan sekitar 85% dari kekhawatiran keselamatan dan kemanjuran yang dicatat oleh FDA dalam pernyataan publik mereka (2). Selain itu, ketika FDA merekomendasikan uji klinis baru, fakta itu ditinggalkan dari komunikasi sponsor pada sekitar 40% kasus.

Surat -surat yang baru dirilis tersedia melalui platform OpenFDA dan dihapus untuk menghapus rahasia dagang atau informasi komersial rahasia (1). Menurut agensi tersebut, hanya CRL yang terkait dengan produk yang disetujui kemudian dimasukkan dalam batch awal ini. FDA sedang meninjau arsipnya dan telah mengindikasikan dapat merilis surat tambahan di masa depan sebagai bagian dari inisiatif transparansi yang lebih luas (1,3).

Tanggapan campuran

Komisaris FDA Marty Makary, MD, MPH, menyatakan bahwa langkah tersebut dimaksudkan untuk mengurangi “permainan menebak” untuk pengembang obat dan investor dan membantu menumbuhkan jalur yang lebih dapat diprediksi untuk persetujuan terapeutik (1).

Kritik terhadap sistem sebelumnya telah lama menunjukkan kurangnya transparansi sebagai faktor dalam kesalahan langkah peraturan berulang di seluruh industri. Ketidakmampuan untuk belajar dari kegagalan orang lain dapat dibilang berkontribusi pada inefisiensi dalam pengembangan klinis dan perencanaan strategis. Publikasi CRLS diharapkan memberi perusahaan dan investor wawasan yang lebih dalam tentang proses pengambilan keputusan agensi dan menyoroti perangkap umum di seluruh domain klinis, manufaktur, dan peraturan (4).

Namun, beberapa pengamat mencatat bahwa rilis menimbulkan pertanyaan tentang konsistensi, waktu, dan ruang lingkup. Misalnya, masih belum jelas apakah CRL di masa depan untuk aplikasi yang tidak disetujui akan dipublikasikan atau apakah sponsor akan memiliki input ke dalam proses redaksi. Analis hukum juga telah menandai pentingnya menjaga perlindungan kerahasiaan sambil memperluas akses ke sejarah peraturan (5).

Sementara reaksi terhadap pergeseran kebijakan telah beragam, publikasi CRLS mewakili perubahan penting dalam bagaimana FDA mengomunikasikan keputusan ulasannya dan dapat menandakan evolusi yang lebih luas dalam standar transparansi peraturan AS.


Referensi

  1. FDA. FDA mencakup transparansi radikal dengan menerbitkan huruf respons lengkap. Siaran pers. 10 Juli 2025.
  2. Downing N, Ross J, dkk. Menjelaskan konten surat tanggapan lengkap FDA dan bagaimana sponsor melaporkannya secara publik: studi cross sectional. BMJ 2015; 350: H2758.
  3. Mason, G.FDA melepaskan 'batch awal' dari lebih dari 200 surat penolakan narkoba. Biotech sengit.
  4. Halpern L. FDA menerbitkan ratusan surat tanggapan lengkap dari paruh pertama dekade ini. Waktu farmasi.
  5. Hogan Lovells. Penciptaan database publik FDA yang terdiri dari 200 CRL menimbulkan pertanyaan. Siaran pers. 10 Juli 2025.

EMA bergerak untuk mengamankan pasokan obat kritis yang digunakan selama kehamilan

Tanda Teks Menampilkan Berita Industri. Teks foto bisnis menyampaikan berita kepada masyarakat umum atau target publik | Kredit Gambar: © Artur – © Artur – Stock.adobe.com

Badan Obat Eropa (EMA) dan Kepala Badan Obat-obatan (HMA) telah mengeluarkan rekomendasi melalui kelompok pengarah eksekutif tentang kekurangan dan keamanan produk obat (MSSG) untuk mengamankan pasokan imunoglobulin anti-D yang digunakan untuk mencegah imunisasi RHD selama kehamilan di Uni Eropa. Pasokan dalam masalah karena jumlah donor plasma telah menurun. Donor dengan imunoglobulin anti-D adalah satu-satunya sumber untuk memproduksi obat-obatan ini, menurut EMA, dan perawatan ini adalah satu-satunya pilihan untuk mencegah imunisasi RHD selama kehamilan.

Kunci takeaways

  • Pasokan imunoglobulin anti-D yang digunakan untuk mencegah imunisasi RHD selama kehamilan berisiko di UE. Jumlah donor yang diimunisasi terbatas dan ada penurunan donor yang ada.
  • Kelompok pengarah kekurangan obat EMA merekomendasikan negara-negara anggota UE untuk membuat rencana untuk mengamankan pasokan imunoglobulin anti-D di UE yang meliputi pertimbangan keselamatan, hukum, etika, dan peraturan.
  • Industri didorong untuk berinvestasi dalam kapasitas manufaktur dan pengembangan alternatif untuk imunoglobulin anti-D yang diturunkan dari plasma.

Imunisasi PhD terjadi, menurut EMA, ketika orang hamil dengan darah RHD-negatif terpapar darah RHD-positif dari janin yang tumbuh dalam tubuh mereka. Reaksi kekebalan yang berpotensi fatal dapat terjadi akibat paparan dan berdampak pada kesehatan janin serta setelah bayi lahir (1).

Kerentanan dalam rantai pasokan

Selama penilaian mereka, MMSG mengidentifikasi kerentanan kunci berikut dalam rantai pasokan (2):

  • Jumlah donor yang diimunisasi terbatas dan ada penurunan donor yang ada.
  • Ada tantangan dalam koleksi, manufaktur, dan pengumpulan batch plasma kecil.
  • Kapasitas global dibatasi oleh sejumlah terbatas pemegang otorisasi pemasaran dan pusat-pusat yang mengumpulkan plasma untuk imunoglobulin anti-D.
  • Ada ketergantungan pada negara -negara di luar UE untuk pasokan plasma untuk produk -produk ini.

Rekomendasi untuk regulator, industri, dan pemangku kepentingan lainnya

MMSG mengeluarkan rekomendasi kepada regulator nasional, Komisi Eropa, industri plasma, dan organisasi penelitian. Rekomendasi baru adalah untuk negara-negara anggota UE untuk membuat rencana untuk mengamankan pasokan imunoglobulin anti-D di UE yang mencakup pertimbangan keselamatan, hukum, etika, dan peraturan. Mengurangi penggunaan yang tidak perlu melalui prosedur seperti skrining prenatal non-invasif harus difokuskan. Perawatan alternatif juga harus diteliti dan dikembangkan. Kampanye komunikasi tentang produk obat yang diturunkan plasma juga harus diimplementasikan.

Rekomendasi juga menyatakan, “Negara -negara Anggota, bekerja sama dengan para ahli, termasuk masyarakat yang terpelajar, organisasi pasien, dan pemangku kepentingan terkait lainnya harus mengembangkan pedoman nasional untuk memfasilitasi prioritas pasien yang memerlukan obat -obatan ini selama situasi kekurangan, jika perlu. MSSG dapat mengoordinasikan pengembangan tingkat prioritas serikat pekerja untuk mengelola kekurangan kritis, terkoordinasi di tingkat serikat” (2).

EMA menyatakan dalam siaran pers bahwa EC harus mengidentifikasi langkah -langkah untuk memastikan pasokan perawatan ini dan berkoordinasi dengan negara -negara anggota UE (1). “Langkah -langkah kebijakan yang ditetapkan dalam Undang -Undang Obat Kritis yang diusulkan dapat dimanfaatkan, seperti pengadaan bersama layanan manufaktur untuk membangun atau meningkatkan pasokan obat -obatan ini ke UE,” kata agen tersebut dalam rilis. Dokumen rekomendasi mengatakan EC juga harus “memfasilitasi kerja sama antara pemangku kepentingan utama, termasuk otoritas darah yang kompeten nasional dan otoritas obat -obatan yang kompeten nasional untuk memastikan koherensi di seluruh kerangka kerja legislatif yang relevan.”

Industri juga didorong untuk memastikan pasokan perawatan ini di Eropa dengan berinvestasi dalam kapasitas manufaktur dan pengembangan alternatif untuk imunoglobulin anti-D yang diturunkan plasma.

Rekomendasi (2) menyatakan bahwa industri plasma dan organisasi penelitian harus “berkolaborasi dengan negara-negara anggota dan Komisi Eropa untuk mengidentifikasi mekanisme yang efektif untuk mendukung pengumpulan dan penggunaan plasma. Pertimbangkan inisiatif dan alat yang diterapkan oleh otoritas pengatur untuk mengamankan pasokan imunoglobulin anti-D dan jika diperlukan, menyediakan data yang relevan yang diperlukan untuk mengidentifikasi dan mengembangkan mekanisme ini.

Referensi

  1. Ema. Memperkuat rantai pasokan imunoglobulin anti-D. Siaran pers. 4 Juli 2025. Https://www.ema.europa.eu/en/news/strenghening-supply-chain-anti-d-immunoglobulins
  2. Ema. Rekomendasi Kelompok Pengarah Eksekutif tentang kekurangan dan keamanan produk obat untuk mengatasi kerentanan rantai pasokan imunoglobulin anti-D. EMA/135603/2025. 23 Juni 2025. Https://www.ema.europa.eu/en/documents/other/recommendations-executive-steering-group-safety-safety-medicinal-products-address-ian–Munoglobulin-supply-chain-vulnerability_en.pdress

Pencernaan Obat: Tren Pembuatan Molekul Besar

Dalam video Digest obat eksklusif ini, para ahli dari beberapa pemain utama di bidang biologi merangkum tren saat ini, mengidentifikasi strategi utama untuk mempercepat pengembangan fase awal, berbagi pendekatan untuk mempercepat jadwal untuk format yang paling kompleks, dan melihat ke masa depan kemajuan dalam pembuatan molekul besar.

Daftar gratis: https://www.pharmtech.com/pt_w/large-molecule-trends

Tinjauan Acara:

Ketika format kompleks mendominasi generasi obat biologis berikutnya, tren dalam pembuatan molekul besar berkembang. Episode ini Pencernaan Narkoba Mengidentifikasi apa tren itu, apa yang saat ini beberapa pendekatan untuk mempercepat jadwal pengembangan tanpa mengorbankan kualitas, bagaimana produsen strategi untuk mempercepat tahap awal pengembangan, dan apa yang akan terjadi di masa depan dalam hal kemajuan generasi berikutnya.

Tujuan Pembelajaran Utama:

  • Apa tren saat ini dalam manufaktur molekul besar
  • Strategi untuk mempercepat pengembangan fase awal sambil mengurangi risiko
  • Pendekatan untuk mempercepat jadwal obat baru yang diselidiki bahkan untuk format yang kompleks
  • Cara untuk menjaga proses CMC tetap fleksibel dan memastikan kualitas produk secara bersamaan

Siapa yang harus hadir:

  • Pengembang Biologi
  • Produsen Biologi
  • Ilmuwan formulasi
  • Ilmuwan pembangunan
  • Peneliti atau inovator dengan fokus pada molekul besar

Pembicara:

Matthew Minakowski
Direktur, Komersial
Pengembangan, mamalia,
Biologi Terpadu
Lonza

Daftar gratis: https://www.pharmtech.com/pt_w/large-molecule-trends

Dosage Form Compounding in the Workplace Environment

Pharmaceutical research and development in laboratory, with focus on medicine preparation, powder substances, and glass vials for drug formulation and testing | Image Credit: © felix_brönnimann – stock.adobe.com

Compounded pharmaceutical dosage forms may be generally described as preparations that are not commercially available from major pharma corporations. These preparations have traditionally been prepared in 503A pharmacies for identified patients; larger quantities of compounded preparations are also prepared by 503B outsourcing facilities for commercial distribution (1). Numerous dosage forms are compounded, including investigational drugs, parenteral nutrition, oncology preparations, radiopharmaceuticals, veterinary medications, and replacement commercial products during drug shortages (2-3). Individual compounded dosage forms may be prepared by pharmacists in community or hospital settings; greater scale compounding may include formulation scientists, engineers, and QA personnel in multi-facility corporations. Compounding may be completely manual or automated and may utilize electronic systems (e.g., automated compounding (ExactaMix), programmable robots (Equashield Pro), or pharmacy management (DoseEdge)). Aside from patients and clients, compounders may have relationships with other compounding organizations; (e.g., 503A hospitals may purchase compounded preparations from 503B facilities, and 503B organizations may provide clinical supplies to pharma industry.

Discussion topics

This discussion describes an ordered process for compounding pharmaceutical dosage forms in workplace environments. The process comprises six stages generally applicable to all compounding (Table I).The first four stages directly address dosage form preparation, evaluation, packaging, and medical information. Stage 5 evaluates and verifies previous completed stages; stage 6 comprises dispensing/shipping to patients or clients. Individual activities within stages will vary depending on the type of compounded dosage form (e.g., sterile vs. non-sterile). Activities in stages may be done by multiple people (e.g., pharmacists, technicians, engineers, etc.).

The compounding process was developed considering academic teaching of compounding theory and practice, problem experiences in multiple workplaces, published regulatory observations, and medication error incidents. Academic compounding typically focuses on placebo formulations and techniques in a classroom or laboratory. Compounding in the workplace is much different—active drugs with specific and sometimes incompatible physicochemical properties, actual patients, emergency circumstances, imperfect facilities, incomplete information, inadequate staffing, excessive workloads, and logistics with interruptions, interferences, and other distractions. Basic activities in respective compounding stages are described; potential problems are identified; technical science, compliance, documentation, and personal responsibility are emphasized. The process builds on pharmaceutical and regulatory concepts, including Quality by Design (QbD) (4-6), Quality Risk Management (QRM) (7), and Pharmaceutical Quality Systems (8,9). Concepts described by professional pharma organizations are included (10-13). The technical responsibilities of management supporting compounding are also addressed.

This discussion assumes organization compliance with USP, federal, state, local, other regulatory, and professional certifications; 503B outsourcing facilities must also comply with pharmaceutical Good Manufacturing Practices (GMP). It also assumes availability of approved policies and standard operating procedures (SOPs) at the site. Non-technical considerations, such as drugs and supplies inventory, third-party reimbursement, personnel development, performance metrics, and other business responsibilities, are not addressed.

Table I. Pharmaceutical Compounding Process

Stage 1 compounding process design

Stage 1 comprises identification of all activities required to completely prepare the compounded dosage form and its associated final package. This includes formulation ingredients, preparation, testing, and other post-compounding activities for dispensing to patients or shipping to clients. Relevant information is integrated in stage 1. When stage 1 is completed, all activities for future performance will have been documented to guide the compounding process. Actions to minimize potential hazards (i.e., what might go wrong) are incorporated in the process. Stage 1 design provides technical and compliance consistency throughout all compounding activities. Stage 1 design activities are executed in stages 2, 3, and 4.

Dosage form identification and characterization. The first significant activity in the compounding process comprises evaluation of the medication order for the compounded dosage form. This includes determining the availability of commercial product fulfilling the medication order and making compounding unnecessary; using a commercial product is preferable to extemporaneous compounding. Compounded dosage forms may not be reimbursed by a third party if similar products are commercially available. If a suitable commercial product does not exist, the compounded dosage form is identified. Objectives analogous to QbD for commercial products, such as Critical Quality Attributes (CQA) and Critical Process Parameters (CPP), are noted. These are straightforward for an IV solution with a single additive; other compounding (e.g., parenteral nutrition, non-sterile formulations, etc.) are much more complex. Clear directions for the dosage form to be compounded are essential.

Compounding information. Concurrent with dosage form identification is verification of information relevant to compounding. Questionable, inconsistent, or otherwise problematic information must be corrected or confirmed. Technical information may be applicable throughout the compounding process (e.g., a light-sensitive drug must be correctly handled during preparation, post-compounding, packaging, shipping, and administration to the patient). Using an investigative approach can be helpful when presented with new compounding—what is known, what is not known and must be determined, and what is the desired outcome? In other words, where do you want to go and what do you need to know and do to get there? This approach will provide guidance and direction for the compounding effort. Missing information has been described as a major cause of medication events (14).

  • Patient information. Patient information, such as patient age, weight, medical diagnosis, hepatic function, kidney function, concurrent drug therapy, and other patient factors affecting formulation and dosage, must be reviewed. Is the medication order consistent with diagnosis (e.g., methotrexate dosage for cancer vs. rheumatoid arthritis)?
  • Drug, excipients, primary package, and dosage. Textbooks with example formulations provide the basis for pharmaceutical compounding (15-17). Drugs and excipients to be used in compounding must be reviewed. FDA published a list of bulk drug substances that can be compounded (18,19); these must comply with USP/NF standards (20-24). Compounding must not include drugs withdrawn for reasons of safety or efficacy (25). Relevant physicochemical properties, solubility, stability, compatibility with excipients, and other factors related to formulation design are reviewed. Commercial products must be clearly identified (e.g., which heparin dosage strength will be used for dilutions (26)). Inactive formulation ingredients may be problematic in certain patients (e.g., lactose intolerance, dye allergy). The primary package for the compounded dosage form may be a polymeric IV bag, plastic syringe, glass bottle, or other container, some of which may be incompatible or require extra precautions (e.g., IV nitroglycerin requires a non-PVC administration set). Difficult-to-compound dosage forms identified by FDA should not be compounded (27). Biologic drugs are not compounded (28).
  • Other technical. Information relevant to labeling, patient instructions, secondary packaging, supply chain shipping (e.g., refrigeration, non-freezing, light protection), and related considerations is assembled. Dosage form labeling is prepared. Medication information for distribution to the patient or to 503B clients is compiled. Assembled information must consider the entire compounding process, including patient use.

Dosage form design and calculations. The compounded dosage form to be prepared is defined based on the medication order and assembled information. Formulation ingredients, process steps, primary package, and relevant information that includes details must be clearly stated in compounding documentation. Exact formulation amounts and process steps that include potential causes of failure are critical. Each medication order should be viewed as a new order and not copied from previous documents; prior calculations must not be assumed to be correct. A specific calculations process has been described (29); calculations may also be incorporated within comprehensive compounding documentation. Calculations must be carefully executed and include written units of measure; omitting descriptive units when calculating is a common cause of error. Problematic notations (e.g., mcg, µg, µ) must be clarified. Calculations should be reviewed by a second person; verification must not be cursory (i.e., the originator is a boss or someone who “never” makes mistakes). Calculations logic, execution, and supporting information must be critically reviewed and include being watchful for number transpositions.

Documentation. The compounding record, work order, batch record, etc. is the permanent record of the compounded preparation retained at the compounding site. Each compounded preparation requires some form of compounding record; a compounded IV solution with a single IV additive prepared according to an SOP will require much less detail than an oral capsule formulation with multiple ingredients and a multi-step compounding procedure. Hospital IV solutions may be recorded within a pharmacy management system or other electronic record (e.g., Epic, DoseEdge). A site SOP should provide documentation templates; a standardized template will increase process uniformity and prevent omissions and errors. The compounding record will be utilized throughout all stages of the compounding process. SOP references are noted for standardized activities; planned deviations are noted in the compounding record. Documentation may contain descriptive information, rationale, ingredients, sources, lot numbers, equipment, process information, references, labeling, patient counseling, and other content; its level of detail must be sufficient to enable an exact repeat preparation of the compounded dosage form. Document clarity is essential; multiple personnel will utilize the compounding record throughout the process. Documentation must not contain drug name abbreviations, problematic units of measure, and numeric problems, such as leading and trailing zeros; awareness of look-alike sound-alike (LASA) drug names (e.g., tetracycline HCl and tetracaine HCl) and other sources of confusion should be prospectively highlighted. Evaluation criteria used post-compounding are identified. A listing of representative compounding record topics is presented in Figure 1. Signature/date of individuals associated with respective content throughout the compounding process is recorded in the compounding record. Compounding records are used for material traceability, adverse event investigations, skills training, and other applications. Some organizations develop a library of master formulation records for frequently prepared dosage forms.

Supporting activities design

  • Stage 2: pre-compounding. Stage 2 activities are also designed in stage 1. These include compounding planning followed by preparation of the compounded dosage form. Precautions for problematic activities are highlighted. Critical process techniques must be reviewed with technicians prior to compounding (e.g., particle size reduction, geometric mixing, levigation, syringe use, etc.); additional training may be necessary. Certain drugs and excipients (e.g., potent drugs, antineoplastics, hormones, peanut oil, allergens, beta-lactams (penicillins)) must not contaminate subsequent compounding and require dedicated or disposable equipment. Personal protective equipment (PPE) may be required for technician protection. Potent and dangerous drugs may require restricted access barrier systems or controlled pressure facilities. Stage 2 activities should be generally described in approved site SOPs; planned deviations are noted in the compounding record.
  • Stage 3: dosage form completion. Stage 3 activities are also designed in stage 1. These include physical evaluation of the dosage form (e.g., appearance, solution clarity, suspension dispersibility, topical cream absence of particulates, color, and other visuals). Dosage form evaluation must be rigorous and consistent with design expectations. Testing is specified in the compounding record; 503B preparations are submitted for analytical laboratory testing, sterility testing, endotoxin, and other GMP requirements; validated test methods are required. The label for the final dosage form is prepared; labeling must be accurate (e.g., codeine vs. codeine phosphate) without spelling or typo errors, especially in the patient’s name. The compounded dosage form is quarantined to prevent release pending dosage form approval in stage 5. Equipment and facilities must be properly cleaned per SOP; cleaning must consider physical properties of ingredients (i.e., hydrophilic vs. hydrophobic). Dedicated equipment must be identified and stored to prevent inadvertent use; use of disposables eliminates equipment cleaning issues. Drugs or ingredients requiring specialized waste disposal are identified. These activities should be described in approved SOPs; additional requirements are added to the compounding record.
  • Stage 4: final package. Stage 4 activities include final requirements for dosage form release to patients, clients, or other recipients. Technical information for patients and clients comprises labeling, dosage, administration, verbal counseling, storage conditions, and other relevant communication. Patient information should be equivalent to that provided by pharma manufacturers for commercial products. The final package provided to the patient/client must ensure correct use of the compounded dosage form. Most activities should be identified in SOPs; deviations are noted in the compounding record. Specialized shipping or storage requirements beyond SOP stipulations are noted.

Stage 1 hazards. Table II provides examples of potential stage 1 hazards, risks, and failure modes (i.e., what might go wrong). Most critical among these are identifying critical dosage form characteristics, assembling and verifying information critical to compounding, calculation errors caused by omitting units of measure in equations, and compounding directions that are too general and non-specific. The point of Table II is to encourage prospective thinking about potential problems; personnel designing the compounded dosage form should anticipate problems and include preventive actions in compounding instructions.

Stage 1 accountability. Signature/date of stage 1 responsible personnel is noted in the compounding record. Personal signature/date affirms responsibility for performance of stage 1 activities.

Figure 1: Example Compounding Record Information

Table II. Potential Compounding Problems in Stage 1

Stage 2 compounding

Stage 2 compounding comprises pre-work and actual preparation of the compounded dosage form per stage 1 directions. Personnel involved in actual compounding must also plan their activities for expeditious performance, especially when compounding sterile dosage forms.

Pre-compounding. Pre-compounding activities include selecting and organizing drugs, materials, equipment, and supplies for efficient dosage form preparation. Personnel who prepare the compounded formulation must have good understanding of compounding techniques before initiating compounding. Activities involved may be conducted in a non-sterile compounding area, laminar air-flow workstation (LAFW) in a clean room environment, or other suitable controlled conditions. Compounding personnel must be dressed appropriately, including scrubs, sterile gowns, gloves, face masks, and personal protective equipment as required.

Training. Technician training on preparatory skills and safety considerations is conducted as needed. Technical skills to prepare compounded dosage forms will vary depending on type and complexity. Sterile dosage form compounding is high risk due to potential contamination. Non-sterile compounding requires familiarity with preparatory techniques for specific dosage forms.

  • Facilities. Preparations containing potent or toxic injectable drugs such as anti-neoplastics may require the use of dedicated clean rooms with differential air pressure monitoring.
  • Equipment and dedicated equipment. Compounding equipment must be carefully examined to ensure cleanliness before use. Certain drugs and excipients may require use of dedicated equipment (e.g., potent drugs, hormones, peanut oil, allergenic materials, beta-lactam (penicillin) drugs, and other ingredients that must not contaminate compounded dosage forms).
  • PPE. PPE beyond usual garb/equipment may be required for technician safety when working with high-alert, oncology, or other hazardous drugs.
  • Material selection and evaluation. Drugs, excipients, and required materials to be used in compounding are assembled. Compounding personnel must be watchful for similar drug names (LASA), standardized packaging, small fonts on labels, and other features potentially contributing to erroneous material selection. Assembled drugs, excipients, and required materials must be critically examined for evidence of stability issues, contamination (e.g., black specks), discoloration, insoluble material in solutions, package integrity, mold growth, expired dating, and other problems (Figure 1). Pictures of final selected materials for inclusion in permanent documentation are recommended.
  • Set-up. Compounding sterile formulations requires aseptic gowning, sterile needles and syringes, alcohol swabs, and other supplies. Figure 2 illustrates a typical arrangement of sterile compounded formulation components in an LAFW. Properly executing planned activities will ensure expeditious preparation of sterile preparations, minimizing potential contamination. Real-time verification of problematic process steps is critical to ensure correct preparation of sterile dosage forms. Contemporaneous observation of sterile compounding through imbedded workstation cameras is recommended; digital photographs and other technology may also be utilized. The Institute for Safe Medication Practices (ISMP) has long discouraged the “syringe pull back method” to verify sterile product compounding (31). Compounding non-sterile formulations includes selection of ingredients, equipment, and other supplies. Equipment must be qualified, clean, and otherwise suitable for use. Figure 3 illustrates a typical arrangement of non-sterile compounded formulation components. Real-time observation of problematic process steps is especially critical to ensure correct preparation of non-sterile compounded dosage forms; video, digital photographs, or other technology are useful for permanent records.

Dosage form preparation. The designed dosage form is then prepared. The formula, compounding procedure, and documentation as designed in stage 1 are executed. Sterile preparations require attention to aseptic techniques; non-sterile preparations must utilize technical processes specific to the individual dosage form.

Stage 2 hazards. Table III provides examples of potential hazards occurring in stage 2. The point of Table III is prospective thinking about potential problems. Most critical among these are insufficient planning of compounding activities, overlooking materials used during compounding necessitating retrieval during compounding, substandard compounding skills (especially with less-frequent or unfamiliar techniques), and aseptic technique “shortcuts.”

Stage 2 accountability. Signature/date of stage 2 responsible personnel is noted in the compounding record. Signature/date affirms personal responsibility for performance of stage 2 activities.

Figure 2. Intralipid 20% with damaged packaging (note black oxygen sensor), undissolved mannitol crystals, and contaminated lactose (black specks).

Intralipid 20% with damaged packaging (black oxygen sensor), undissolved mannitol crystals, and contaminated (black specks) lactose.


Figure 3. Sterile Product Compounding Planning

Figure 3. Sterile Product Compounding Planning

Figure 4. Non-Sterile Product Compounding Planning

Figure 4. Non-Sterile Product Compounding Planning

Table III. Potential Compounding Problems in Stage 2


Stage 3 dosage form completion

Stage 3 post-compounding comprises testing, other dosage form-related activities, and ancillary activities executed after completion of compounding.

Post compounding. Activities directly related to the compounded dosage form described in site SOPs are completed. Deviations to SOPs are identified in the compounding record. Most important among these are the critical evaluation of the compounded preparation, submission of samples for testing, and completion of compounding documentation.

  • Sampling and testing. The compounded preparation is evaluated. Visual examination of appearance, color and clarity of solutions, suspension dispersability, topical cream smoothness, absence of solids, freedom from visible foreign particulate matter, package integrity, signs of instability, and other quality attributes are evaluated; physical observation evaluation must be rigorous. 503B compounding samples are submitted for analytical testing to verify potency, sterility, stability, and other requirements identified in stage 1; testing must use validated test methods.
  • Technical review. Deviations, non-conformances, environmental monitoring, and other data related to the compounded dosage form are reviewed. Acceptable results are required for eventual dispensing to patients and shipping to clients.
  • Documentation. Compounding record documentation is completed. Compounding documentation must ensure full traceability of ingredients, processes, and equipment, and it must provide sufficient detail to enable an identical repeat preparation of the compounded dosage form. Documentation for sterile preparations is completed outside the LAFW.
  • Labeling. The compounded dosage form is labeled and associated technical information assembled per stage 1 directions.
  • Quarantine. The compounded dosage form is quarantined until all testing is completed and reviewed.

Ancillary activities. Ancillary activities include workspace and equipment cleaning and waste disposal. If dedicated equipment is used for specific ingredients, it must be cleaned, dried, labeled, and appropriately stored to prevent future inadvertent use; misuse of dedicated equipment is a common compounding error. Waste disposal must follow site procedures (i.e., “sharps,” recyclables, HIPAA, biologicals, oncology drugs, and drug waste disposed into appropriate containers). Routine post-compounding activities are completed per SOPs. Deviations from SOPs are noted in the compounding record.

Stage 3 hazards. Examples of possible stage 3 hazards are presented in Table IV. The point of Table IV is prospective thinking about potential problems. A common stage 3 problem is dosage form evaluation; preparations must strive for excellence and not be just “good enough.” Equipment used in compounding must be scrupulously cleaned after use. Dosage forms must be quarantined to prevent release until final approval (stage 5),

Stage 3 accountability. Signature/date of stage 3 responsible personnel is recorded in the compounding record. Signature/date affirms personal responsibility for stage 3 activities.

Table IV. Potential Compounding Problems in Stage 3

Stage 4 final package

Stage 4 final package comprises technical and associated activities that complete dosage form preparation. Supportive information for patients or clients is compiled as specified in stage 1.

Technical. Test results are compiled and evaluated. Failing or unexpected test results must be investigated, appropriate action taken, and corrective action implemented. Labeling, medication information, patient counseling, and other communication to be provided to patents and clients is compiled. Commercial product manufacturers provide medication information to patients; comparable information should be developed on patient/client interactions. ISMP Medication Learning Guides for Consumers exemplify drug information provided to patients at dispensing (34). The compounded dosage form must be used correctly by the patient to ensure efficacy.

Associated activities are identified in an SOP or specified in the compounding record. Deviations to SOPs may include specialized protective packaging, time/temperature limitations with tracking devices, and other shipping requirements. Shipping should be performed by a dedicated delivery service in a controlled manner with location-tracking capability; shipping must include temperature control.

Documentation. Compounding record documentation is finalized. This is the final compounding record retained by the organization. After the compounded dosage form is dispensed or shipped, the compounding record is the lasting evidence of compounded dosage form preparation.

Stage 4 hazards. Examples of possible stage 4 hazards are presented in Table V. The point of Table V is prospective thinking about potential problems. Most important among these are assembly of communication for patients and clients. The compounded dosage form must be used properly by the patient, administered correctly, and otherwise handled to be efficacious. Completion of documentation is also critical in stage 4. Compounded dosage form quarantine (stage 3) should prevent inadvertent release to patients or clients.

Stage 4 accountability. Signature/date of stage 4 responsible personnel is recorded in the compounding record. Signature/date affirms personal responsibility for stage 4 activities.

Table V. Potential Compounding Problems in Stage 4

Stage 5 verification

Stage 5 verification is the final check on the compounded dosage form, packaging, documentation, and related information before dispensing to the patient or shipping to clients.

Review and evaluation. Review of the completed dosage form and associated elements is a critical step in the compounding process. Ideally, verification is accomplished by an independent observer separate from people involved in the compounding process. Verification addresses the final dosage form, test results, completed documentation, and associated communications to be provided to patients or clients. The compounded dosage form is not dispensed to patients or shipped to clients until the final compounded dosage form and associated information are approved.

Review by a competent, independent observer must be a serious and thorough activity by an impartial observer—not a perfunctory ”rubber stamp” exercise. ISMP has discussed the judicious use of independent double-checks in pharmacy practice (35). Final dosage form and associated information must be reviewed vs. the original medication order. Review must be completed before the compounded dosage form is dispensed or shipped. Checklists are useful to ensure a comprehensive review. Errors, omissions, or insufficient details in compounding documentation are corrected. Documentation will be utilized in future investigations, training, and other applications. Final documentation is retained in prescription files, patient records, manufacturing batch records, or electronic systems. An example checklist with relevant topics is provided in Figure 5.

Compounded dosage form approval. When dosage form appearance, test results, documentation, and other reviewed content are acceptable, the compounded dosage form is approved for release by the responsible pharmacist. Approval is documented in the compounding record. The dosage form is removed from quarantine.

Stage 5 hazards. Examples of possible stage 5 verification hazards are presented in Table VI. The point of Table VI is prospective thinking about potential problems. Potential hazards in stage 5 are critical; this is the final check of dosage form quality, test results, patient/client documentation, and compounding record documentation. Compounding record documentation is the lasting record of the compounding process.

Stage 5 accountability. Signature/date of stage 5 responsible personnel is recorded in the compounding record. Signature/date affirms personal responsibility for stage 5 activities.

Figure 5. Example Compounding Verification Checklist

Table VI. Potential Compounding Problems in Stage 5

Stage 6 dispensing/distribution

Stage 6 comprises activities in which compounded dosage forms are provided to users.

Dispensing/shipping to patients/clients. Stage 6 includes dosage form dispensing to patients, providing drug information, and counseling as needed. Dispensing and counseling are performed according to the clinical situation and patient needs. Stage 6 also includes shipping to patients per secondary package design, including temperature control, light protection, package tracking, and other requirements. Approval to dispense/ship is documented in stage 5; dispensing or shipping must not occur before the responsible pharmacist has provided final dosage form approval. Stage 6 also includes shipping to clients per secondary package design after responsible pharmacist approval.

Numerous examples exist of drug products being dispensed to the wrong patient and conditions under which this may occur. Errors may occur in community settings or in the hospital environment for a variety of reasons. Obvious causes involve patients with identical last names. Hospitalized patients may switch beds without notifying the nursing staff. Electronic order entry may be confused; mis-transcribed telephone orders, dispensing system overrides, and other potential patient mixups may occur. Verification using two patient identifiers is recommended to confirm the intended patient as well as verify correct treatment to the individual. These actions should occur at each step in the medication process (i.e., from original medication order, through multiple transcriptions {physicians, pharmacists, nurses}, and final dosage form administration to the patient). Examples of questions to be asked before drug administration, dispensing, or counseling to verify they are the intended patient typically include the patient’s name, date of birth, last four digits of the US social security number, or other government identification. Only verifying the patient’s name is not sufficient. Patients for whom English is a second language require increased assurance that they are the intended patient.

Shipping to clients. Stage 6 also includes shipping to clients per secondary package design. Approval to dispense/ship is documented in Stage 5; again, dispensing or shipping must not occur before the responsible pharmacist has provided final dosage form approval.

Stage 6 hazards. Examples of possible stage 6 hazards are presented in Table VII. The point of Table VII is prospective thinking about potential problems. Most significant among these are dispensing a drug to the wrong patient. This may occur in community and hospital settings; there are numerous opportunities for actions that contribute to dispensing/administration of dosage form to the wrong patient (36-38).

Stage 6 accountability. Signature/date of stage 6 responsible personnel is noted in the compounding record.Signature/date affirms personal accountability for stage 6 activities.

Table VII. Potential Compounding Problems in Stage 6

Management responsibilities

Management of a compounding organization is responsible for daily compounding performance, as well as related supportive activities. Quality systems guidance documents identify expectations of management (e.g., leadership, structure, quality system design, policies, objectives, plans, system review). FDA issued CDER’s Quality Management Maturity Program (39) addressing expectations beyond current GMP; compounding management must aspire to much more than minimal GMP compliance. Development of a quality culture mindset in which risks to compounded product quality are minimized is led by management (40)and should be forefront in the organization. ICH Q9(R1) QRM addresses “What might go wrong?” and describes common methods of risk assessment; these methods are directly applicable to pharmaceutical compounding.

Primary management responsibilities in compounding organizations include the following:

  • Personnel. Hiring capable personnel is the most important function of management. Compounding personnel must have appropriate technical compounding skills; organizational skills, self-discipline, attention to detail, language skills, legible handwriting, common sense, and realization of personal deficiencies (i.e., knowing what you don’t know) are desirable. Interpersonal skills in a teamwork environment are essential. Sufficient number of personnel must also be hired commensurate with workload.Management is also responsible to assure that each staff member knows and understands their roles and responsibilities, as well as accepts personal responsibility for performance.
  • Plans, policies, and procedures. Management is responsible for the standards that define daily workplace activities. Compounding organizations must have written and approved documents; compliance with these documents is mandatory. Inadequate procedures, training, and non-compliant performance are always among the most frequent GMP violations in pharma (41). SOP compliance must be forefront in compounding organization performance. SOPs should be living documents, periodically reviewed for accuracy and relevance and updated when new information is available.
  • Training. Personnel must be trained and periodically re-trained on SOPs. Training must be documented; if not documented, it didn’t happen. Training must emphasize personal accountability for SOP requirements; personnel must understand that their signature/date in compounding documentation affirms personal responsibility.
  • Communication. The workplace environment must encourage communication of problems, mistakes, and issues without fear of punishment or embarrassment. Management must develop a collaborative relationship with staff with ongoing focus on quality (i.e., a true quality culture that is evident in personnel performance). ISMP identified communications about medication safety risks occurring within and outside the organization as best practices for community (42) and hospital pharmacy (43). FDA-483 observations and warning letters demonstrating compliance deficiencies in compounding organizations are readily available (44).
  • Commitment. Beyond hiring, procedures, and training are the everyday expressions of management commitment to quality. Management focus must expand from traditional management to include anticipation of performance hazards and risks (i.e., what might go wrong). Management daily performance in routine activities must exemplify commitment to quality.
  • Administrative. Management is responsible for administrative functions supportive to compounding (e.g., funding, scheduling, maintenance, calibration, facility cleanliness).

Management hazards.Examples of possible management hazards are presented in Table VIII. Most significant among these are deficiencies in management commitment to responsibilities; organization culture is directly related to management commitment. The actions of individual employees reflect the attitude and commitment of management—what management wants, management gets. Management is also ultimately responsible to address potential hazards identified above in respective stages of the compounding process.

Management accountability. Signature/date of management on policies, procedures, and other documents affirms management accountability.

Table VIII. Potential Compounding Problems by Management

Summary

This discussion describes an ordered process for pharmaceutical compounding based on academic teaching of compounding theory and practice, problem experiences in multiple settings, regulatory observations, and published medication errors. Technical activities occurring in the compounding workplace beyond physical compounding techniques are addressed. The process describes what and why activities, as well as what might go wrong throughout the comprehensive process.

Six stages of compounding activities are identified, each having specific objectives. Stage 1 design activities address considerations to be executed in all subsequent compounding stages; technical consistency throughout the process is forefront. Compounding activities are executed in stages 2, 3, and 4. Stage 2 comprises selection of materials and equipment, compounding planning, and actual dosage form preparation. Stage 3 post-compounding comprises dosage form review, sampling, testing, and related activities. Stage 4 includes final review of test results; medical information and packaging for dispensing to patients or shipping to clients is compiled. Stage 5 verification comprises review of the compounded dosage form, labeling, drug information, and secondary packaging; stage 5 activities require an independent observer. The compounded medication is approved for dispensing in stage 5. Stage 6 dispensing/distribution provides the compounded dosage forms to users, including patient counseling, medical information, and/or appropriate supply chain shipping. Compliance with regulations and site procedures is required throughout the process. Personnel understanding of their responsibilities and affirming accountability by signature/date on documentation is required. Examples of potential hazards are identified throughout; case studies further demonstrate problematic circumstances.

Management has significant technical responsibilities in the compounding process. Management is responsible for the actual compounding, as well as for supportive functions. Management must continually support employee training and awareness of possible problems. As risks are identified and knowledge is gained, SOPs are modified, including corrective and preventive actions for continuous improvement. A workplace environment in which quality is forefront (i.e., a quality culture) must be an ongoing goal of compounding management and staff.

References

  1. Pluta, P.L. Pharmaceutical Compounding Overview – Technical, Regulatory, and Management in a Workplace Environment. Pharmaceutical Technology 2024, 48(2) 18-22. Compounding Overview: Primary Considerations for the Workplace (pharmtech.com). (accessed 7-1-25)
  2. FDA. Drug Shortages, https://www.accessdata.fda.gov/scripts/drugshortages/default.cfm (accessed 7-20-24).
  3. FDA. Compounding Certain Ibuprofen Oral Suspension Products Under Section 503B of the Federal Food, Drug, and Cosmetic Act. February, 2023. https://www.fda.gov/media/164693/download (accessed 7-1-25).
  4. Yu, L. X. Pharmaceutical Product Quality, Quality by Design, cGMP, and Quality Metrics. FDA. 01-yu-fda-pqri-2015-ver1.pdf (accessed 7-1-25).
  5. Yu, L.X., G. Amidon, M.A. Khan, S.W. Hoag, J. Polli, G.K. Raju, and J. Woodcock. Understanding Pharmaceutical Quality by Design. MIT Open Access Articles. AAPS J 16, 34, 5-23-2014. 771-783. https://dspace.mit.edu/bitstream/handle/1721.1/106646/12248_2014_Article_9598.pdf?sequence=1 (accessed 7-1-25).
  6. ICH Q8(R2). Pharmaceutical Development. August 2009. Q8_R2_ (ich.org) (accessed 7-1-25).
  7. ICH Q9(R1). Quality Risk Management, Q9(R1) 1-18-2023. https://database.ich.org/sites/default/files/ICH_Q9%28R1%29_Guideline_Step4_2023_0126_0.pdf (accessed 7-1-25).
  8. FDA. Guidance for Industry.Quality Systems Approach to Pharmaceutical CGMP Regulations, September, 2006. https://www.fda.gov/media/71023/download (accessed 7-1-25).
  9. ICH. Pharmaceutical Quality System Q10. 6-4-2008. https://database.ich.org/sites/default/files/Q10%20Guideline.pdf (accessed 7-1-25).
  10. American Society of Health System Pharmacists. ASHP Guidelines on Compounding Sterile Preparations https://www.ashp.org/-/media/assets/policy-guidelines/docs/guidelines/compounding-sterile-preparations.ashx (accessed 7-1-25).
  11. National Association of Pharmacy Regulatory Authorities. Guidance Document for Pharmacy Compounding of Non-Sterile Preparations, 2018. NAPRA-Mdl-Stnds-Pharmacy-Compounding-Nonsterile-Preparations-Guidance-EN-June-2018-CLAR-Jan-2022.pdf (accessed 7-1-25).
  12. ISMP. ISMP Guidelines for Safe Preparation of Compounded Sterile Preparations, 2016. https://www.ismp.org/sites/default/files/attachments/2017-11/Guidelines%20for%20Safe%20Preparation%20of%20Compounded%20Sterile%20Preperations_%20revised%202016.pdf (accessed 7-1-25).
  13. Campbell, Robert. The Joint Commission’s Medication Compounding Certification Program, 2017. https://www.pppmag.com/article/2068#:~:text=Campbell%3A%20The%20Joint%20Commission%20Medication%20Compounding%20Certification%20is,%3C797%3E.%20Certification%20is%20not%20offered%20for%20outsourcing% (accessed 7-20-24).
  14. Shaw, Gina. Missing Information a Top Cause of Medication Events. Pharmacy Practice News, 7-26-24. https://www.pharmacypracticenews.com/Operations-and-Management/Medication-Safety/Article/07-24/Missing-Information-a-Top-Cause-of-Medication-Events/74203 (accessed 7-1-25).
  15. Allen, Lloyd V., Jr. The Art, Science, and Technology of Pharmaceutical Compounding, 6th ed. American Pharmacists Association, Washington, DC, 2020.
  16. Allen, Lloyd V, Jr. International Journal of Pharmaceutical Compounding. https://ijpc.com/Index.cfm (accessed 7-1-25).
  17. Shrewsbury, Robert. P. Applied Pharmaceutics in Contemporary Compounding, 3rd ed. Morton Publishing, Englewood, CO 80110) 2015.
  18. FDA. 21 CFR §216.23. Bulk drug substances that can be used to compound drug products in accordance with section 503A of the Federal Food Drug, and Cosmetic Act. https://www.ecfr.gov/current/title-21/chapter-I/subchapter-C/part-216/subpart-B/section-216.23 (accessed 7-1-25).
  19. FDA. 503B Bulks List. 8-21-2023. (accessed 7-20-24)
  20. USP Pharmaceutical Compounding — Non-Sterile Preparations, 1-11-22. https://www.usp.org/compounding/general-chapter-795 (accessed 7-1-25).
  21. USP Pharmaceutical Compounding — Sterile Preparations, 1-11-22. https://www.usp.org/compounding/general-chapter-797 (accessed 7-1-25).
  22. USP Hazardous Drugs – Handling in Healthcare Settings. https://www.usp.org/compounding/general-chapter-800 (accessed 7-1-25).
  23. USP Quality assurance in Pharmaceutical Compounding. https://www.drugfuture.com/Pharmacopoeia/USP32/pub/data/v32270/usp32nf27s0_c1163.html#google_vignette (accessed 7-1-25).
  24. USP Compounding Compendium. https://www.usp.org/products/usp-compounding-compendium (accessed 7-1-25).
  25. FDA. 21 CFR §216.24. Drug products withdrawn or removed from the market for reasons of safety or effectiveness. https://www.ecfr.gov/current/title-21/chapter-I/subchapter-C/part-216/subpart-B/section-216.24 (accessed 7-1-25).
  26. Arimura, J., R.L. Poole, M. Jeno, W. Rhine, and P. Sharek. Neonatal Heparin Overdose—A Multidisciplinary Team Approach to Medication Error Prevention. J.Pediatr Pharmacol Ther. 2008 Apr-Jun 13(2); 96-98. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3462065/ (accessed 7-1-25).
  27. FDA. Drug Products or Categories of Drug Products that Present Demonstrable Difficulties for Compounding under Sections 503A or 503v of the federal Food, Drug, and Cosmetic Act, 3-20-24. https://www.federalregister.gov/documents/2024/03/20/2024-05801/drug-products-or-categories-of-drug-products-that-present-demonstrable-difficulties-for-compounding (accessed 7-1-25).
  28. FDA. Compounding and the FDA: Questions and Answers. 6-29-22. https://www.fda.gov/drugs/human-drug-compounding/compounding-and-fda-questions-and-answers#:~:text=Can%20biologics%20be%20compounded%3F%20No.%20Biological%20products%20are,the%20scope%20of%20an%20approved%20biologics%20license%20application (accessed 7-1-25).
  29. Pluta, P.L., A. Mancini, N.B. Thakar, and V. Chaiyaperm. Dosage Form Calculations Process in a Workplace Environment. Pharmaceutical Technology. Trends in Manufacturing ebook, 5-2024, 22-34. https://www.pharmtech.com/view/pharmaceutical-compounding-calculations-in-a-workplace-environment (accessed 7-1-25).
  30. ISMP. Life-threatening Errors with Flecainide Suspension in Children. 4-23-2015. https://www.ismp.org/sites/default/files/attachments/2018-03/community201504.pdf (accessed 7-1-25).
  31. ISMP. Maximize Benefits of IV Workflow Management Systems by Addressing Workarounds and Errors. 9-7-17. https://www.ismp.org/resources/maximize-benefits-iv-workflow-management-systems-addressing-workarounds-and-errors (accessed 7-1-25).
  32. ISMP. Too Close for Comport; Fatal zinc overdose narrowly avoided. 7-4-19. https://www.nutritioncare.org/uploadedFiles/Documents/Newsletter/ISMP%20Newsletter%20July%204%202019%20-%20near%20miss%20zinc%20overdose.pdf (accessed 8-1-24).
  33. Grissinger, Matthew. A Fatal Zinc Overdose in a Neonate. P&T, Jul 36(7), 2011. https://ncbi.nlm.nih.gov/pmc/articles/PMC3171817/ (accessed 7-1-2).
  34. ISMP. High-Alert Medication Learning Guides for Consumers, 11-2-18. https://www.ismp.org/resources/high-alert-medication-learning-guides-consumers (accessed 7-1-25).
  35. ISMP. Independent Double Checks: Worth the Effort if Used Judiciously and Properly. 6-6-2019. https://www.ismp.org/resources/independent-double-checks-worth-effort-if-used-judiciously-and-properly (accessed 5-1-24).
  36. Yang, Annie and M. Grissinger. Wrong-Patient Medication Errors: An Analysis of Event Reports in Pennsylvania and Strategies for Prevention. Pennsylvania Patient Safety Advisory, Vol 10, #2, June 2013. https://patientsafety.pa.gov/ADVISORIES/Documents/201306_home.pdf (accessed 7-1-25)
  37. Schulmeister, Lisa. Patient Misidentification in Oncology Care. Clinical Journal of Oncology Nursing, V 12, #3, June 2008. https://pubmed.ncbi.nlm.nih.gov/18515248/ (accessed 7-1-25).
  38. Grissinger, Matthew. Oops, Sorry, Wrong Patient. P&T 39(8), 535-537, Aug 2014. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4123800/ (accessed 7-1-25).
  39. FDA. CDER Quality Management Maturity. 1-26-24. https://www.fda.gov/drugs/pharmaceutical-quality-resources/cder-quality-management-maturity#:~:text=FDA%E2%80%99s%20Center%20for%20Drug%20Evaluation%20and%20Research%20%28CDER%29,go%20beyond%20current%20good%20manufacturing%20practice%20%28CGMP%29%20requirements (accessed 7-1-25).
  40. Friedman, Rick. Establishing a Culture of Quality, September, 2021. https://www.fda.gov/media/156357/download?attachment (accessed 7-1-25)
  41. Hoare, Aaron. The Most Common FDA 483 Observations. https://www.ideagen.com/thought-leadership/blog/the-most-common-fda-483-observations, 4-18-2023 (accessed 7-1-25).
  42. ISMP. First ISMP Targeted Medication Safety Best Practices for Community Pharmacy Released, 4 Apr, 2023. https://www.ismp.org/news/first-ismp-targeted-medication-safety-best-practices-community-pharmacy-released (accessed 7-20-24).
  43. ISMP. Best Practice 14 in 2018-2019 Targeted Medication Safety Best Practices in Hospitals. https://www.ismp.org/sites/default/files/attachments/2019-01/TMSBP-for-Hospitalsv2.pdf (accessed 7-1-25).
  44. FDA. Compounding: Inspections, Recalls, and Other Actions, 4-23-24. https://www.fda.gov/drugs/human-drug-compounding/compounding-inspections-recalls-and-other-actions (accessed 7-1-25).

Acknowledgments

Contributions from Jan M. Keresztes, RPh, PharmD, pharmaceutical educator, Jeanne Moldenhauer, pharmaceutical consultant, Excellent Pharmaceutical Consultants, Richard Poska, RPh, managing director, Flexo CMC Consulting, William R. Porter, PhD, principal scientist, Peak Process Performance Partners, and Chloe Gutierrez, student at Roosevelt University College of Science, Health, and Pharmacy, Schaumberg, IL, are greatly appreciated.

Authors

Paul L. Pluta, RPh, PhD, is a pharmaceutical scientist with pharmaceutical industry, academic teaching, journal editorship, community pharmacy, and hospital pharmacy experience.

Alan M. Mancini, RPh, is a pharmaceutical scientist with pharmaceutical industry, academic teaching, community pharmacy, and hospital pharmacy experience.

Nishant B. Thakar, RPh, PharmD, is Assistant Professor Clinical Sciences, Roosevelt University College of Science, Health, and Pharmacy, Schaumburg, IL, USA, with community pharmacy and hospital pharmacy experience.

Varanya Chaiyaperm, RPh, PharmD is Clinical Assistant Professor University of Illinois College of Pharmacy, Chicago, IL, USA, with community pharmacy and hospital pharmacy experience.

Perawatan theramex untuk osteoporosis pada wanita pascamenopause yang diterima oleh konsorsium obat -obatan Skotlandia

Struktur Tulang Ilustrasi 3D, normal dan dengan osteoporosis | Kredit Gambar: © Adimas – Stock.adobe.com

Konsorsium Obat-obatan Skotlandia (SMC), melalui Layanan Kesehatan Nasional (NHS) di Skotlandia, telah merekomendasikan abaloparatide, yang dipasarkan oleh Theramex yang berbasis di London sebagai Eladynos, untuk pengobatan osteoporosis pada wanita pascamenopause yang berisiko sangat tinggi (1). Theramex dan SMC keduanya membuat pengumuman pada 7 Juli 2025, dengan SMC juga menerbitkan saran tentang tujuh obat lain (1,2).

Statistik menggelegar untuk wanita yang terkena dampak

Kunci takeaways

  • SMC telah merekomendasikan ELADYNOS THERAMEX (Abaloparatide) untuk wanita pascamenopause di Skotlandia dengan risiko tinggi patah tulang, memperluas akses di seluruh Inggris.
  • Dengan fraktur pinggul yang menghitung NHS £ 2 miliar (US $ 2,7 miliar) per tahun, penggunaan perawatan osteoporosis yang lebih luas seperti abaloparatide dapat membantu mengurangi biaya perawatan kesehatan jangka panjang.
  • SMC juga menyetujui gamma bevacizumab, osimertinib, pembrolizumab, dan selpercatinib untuk berbagai kanker, sementara menurun tiga terapi profil tinggi lainnya.

Dalam siaran pers, Theramex memperkirakan bahwa lebih dari 1000 wanita pascamenopause di Skotlandia akan memenuhi syarat untuk perawatan, dengan rekomendasi SMC mengikuti yang sebelumnya dibuat pada Agustus 2024 oleh Institut Nasional untuk Kesehatan dan Perawatan Keunggulan (Nice) yang mengatur status Abaloparatide di Inggris, Wales, dan Irlandia Utara (1). Theramex mengatakan diperkirakan bahwa setengah dari semua wanita di atas 50 akan mengalami fraktur kerapuhan, yang dibuat lebih mungkin oleh osteoporosis, dengan fraktur pinggul bertanggung jawab sekitar 50% kematian terkait fraktur pada wanita.

Seperempat pasien yang menderita patah tulang pinggul mati dalam waktu satu tahun, kata Theramex, dengan patah tulang pinggul seharga NHS £ 2 miliar (US $ 2,7 miliar) per tahun dan fraktur osteoporotik saat ini menyumbang lebih dari £ 4,6 miliar (US $ 6,3 miliar) dari biaya langsung TO NHS, perkiraan peningkatan £ 6 miliar.

“Dengan wanita dengan risiko osteoporosis lima kali lebih besar daripada pria, saya senang bahwa kami dapat memperluas akses lebih jauh di seluruh (Inggris) dan membawa perawatan ini ke wanita di Skotlandia,” Tina Backhouse, manajer umum Inggris di Theramex, mengatakan dalam rilisnya (1). “Persetujuan ini tidak hanya memastikan wanita di Skotlandia memiliki akses ke beragam pilihan perawatan tetapi juga memperkuat upaya berkelanjutan kami untuk berkolaborasi dengan penyedia layanan kesehatan dan pembuat kebijakan untuk menghilangkan perbedaan dalam perawatan osteoporosis di seluruh Inggris.”

Persetujuan SMC, ketidaksetujuan mencakup jangkauan yang luas

Pendapat positif lainnya yang diterbitkan oleh SMC dalam rilisnya 7 Juli termasuk yang berikut:

  • Bevacizumab Gamma (Lytenava, Outlook Therapeutics) diterima untuk pengobatan orang dewasa dengan degenerasi makula terkait usia basah.
  • Osimertinib (Tagrisso, AstraZeneca) diterima, bersama dengan kemoterapi, untuk pengobatan lini pertama orang dewasa dengan kanker paru-paru sel non-kecil di mana sel kanker memiliki mutasi pada ekson 19 atau ekson 21 gen EGFR.
  • Pembrolizumab (Keytruda, Merck-dikenal sebagai MSD di luar Amerika Serikat dan Kanada) diterima, bersama dengan kemoterapi, untuk pengobatan lini pertama orang dewasa dengan kanker endometrium lanjut yang baru didiagnosis atau berulang.
  • Selpercatinib (Retsevmo, Eli Lilly and Company), diterima untuk pengobatan pasien berusia 12 dan lebih tua dengan kanker tiroid lanjut yang disebabkan oleh mutasi pada gen RET (2).

Sebaliknya, SMC menolak merekomendasikan obat -obatan berikut:

  • Amivantamab (Rybrevant, Johnson & Johnson), bersama dengan kemoterapi, tidak direkomendasikan untuk pengobatan orang dewasa dengan kanker paru-paru sel non-kecil di mana sel kanker memiliki mutasi pada ekson 20 gen EGFR.
  • Fezolinetant (Veoza, Astellas) tidak direkomendasikan untuk pengobatan gejala vasomotor sedang hingga berat yang disebabkan oleh menopause, seperti hot flash dan keringat malam.
  • Lecanemab (Leqembi, Eisai/Biogen) tidak direkomendasikan untuk pengobatan penyakit Alzheimer stadium awal pada orang dewasa yang membawa satu atau tidak ada salinan gen APOE4 (2).

Sebelumnya pada tahun 2025, NICE memberikan pendapat positif pada bulan Januari untuk pengobatan Osimertinib AstraZeneca untuk pengobatan pasien kanker paru -paru di Inggris dan Wales; Pada bulan Februari, Komite Badan Obat Eropa untuk Produk Obat untuk Penggunaan Manusia menguatkan keputusan aslinya tentang perpanjangan indikasi untuk pembrolizumab Merck, dan mengatakan otorisasi pemasaran untuk EISAI dan Lecanemab Biogen tidak perlu diperbarui (3,4).

Referensi

1. THERAMEX. SMC merekomendasikan Eladynos (abaloparatide) untuk mengobati osteoporosis pada wanita pascamenopause. Siaran pers. 7 Juli 2025.
2. Konsorsium Obat -obatan Skotlandia. Rilis Berita Keputusan Juli 2025. Siaran pers. 7 Juli 2025.
3. AstraZeneca. AstraZeneca menerima dua rekomendasi bagus positif untuk pasien kanker paru -paru di seluruh Inggris dan Wales. Siaran pers. 22 Januari 2025.
4. EMA. Sorotan pertemuan dari Komite untuk Produk Obat untuk Penggunaan Manusia (CHMP) 24-27 Februari 2025. Siaran Pers. 28 Februari 2025.

Sandoz mengenang cefazolin karena kesalahan pengemasan, membuka fasilitas biosimilar di Slovenia

Tabung Uji Laboratorium Sains, Peralatan Laboratorium | Kredit Gambar: © miliarphotos.com – © miliarphotos.com – stock.adobe.com

FDA yang diumumkan pada tanggal 27 Juni 2025, bahwa Sandoz, Inc. secara sukarela mengingat satu lot cefazolin untuk injeksi, USP, 1 gram per botol setelah pelanggan mengeluh bahwa empat kalium penisilin untuk injeksi, USP, 20 juta unit botol) tidak benar termasuk dalam karton Cefazolin untuk cetak vial. Sementara perusahaan menyatakan belum menerima laporan tentang efek samping yang terkait dengan penarikan, mereka memang menerima satu keluhan tentang produk yang salah yang diberikan kepada pasien.

Kunci takeaways

  • Empat botol kalium penisilin g untuk injeksi, USP salah dimasukkan dalam banyak cefazolin untuk injeksi, USP.
  • Kedua antibiotik memiliki indikasi yang berbeda. Menggunakan kalium penisilin untuk injeksi, USP alih -alih cefazolin untuk injeksi, USP bisa mengancam jiwa.
  • Fasilitas baru Sandoz di Slovenia akan menghasilkan produk yang dapat disuntikkan untuk biosimilar yang ada dan yang akan datang.
  • Fasilitas baru ini merupakan bagian dari investasi $ 1,1 miliar yang telah direncanakan perusahaan hingga 2029.

Nomor lot yang terkena dampak adalah PG4360, nomor Vial National Drug Code (NDC) 0781-3451-70, dengan tanggal kedaluwarsa 2027-Nov (1). Lot diproduksi oleh Sandoz GmbH dan didistribusikan oleh Sandoz Inc. Botol yang salah termasuk memiliki vial NDC 0781-6136-94 dan tanggal kedaluwarsa 2027-nov.

Dua antibiotik memiliki indikasi yang berbeda

Cefazolin untuk injeksi USP diindikasikan untuk pengobatan dan/atau pencegahan infeksi bakteri di berbagai bagian tubuh pada orang dewasa, orang tua, dan anak -anak (1). Penicillin G potassium for injection treats certain serious infections, such as septicemia, skin infections, and wound infections, and can be used to treat diphtheria, community-acquired pneumonia, peritonitis, meningitis/brain abscesses, osteomyelitis, infections of the genital tract, anthrax, tetanus, gas gangrene, listeriosis, pasteurellosis, rat bite fever, fusospirochetes, actinomycosis, komplikasi pada gonore dan sifilis, dan lyme. Ini harus digunakan untuk hanya mengobati atau mencegah infeksi yang disebabkan oleh bakteri yang rentan untuk mengurangi kemungkinan bakteri yang resistan terhadap obat berkembang.

Menurut siaran pers FDA (1), kedua antibiotik memiliki indikasi yang berbeda meskipun menjadi bagian dari kelompok antibiotik beta-laktam yang sama dan memiliki populasi pasien yang tumpang tindih. Regimen dosis untuk masing -masing mungkin juga berbeda.

Pemberian injeksi kalium penisilin g alih-alih injeksi cefazolin mungkin memiliki konsekuensi yang mengancam jiwa yang mencakup kurangnya kemanjuran yang mengakibatkan pengobatan infeksi parah yang kurang optimal, resistensi antibiotik, reaksi alergi, interaksi obat, dan aritmia jantung yang disebabkan oleh kalium tinggi.

Sandoz meminta pelanggan untuk mengembalikan produk yang ditarik, yang dikirim untuk memilih pedagang grosir untuk distribusi nasional. Perusahaan merekomendasikan penyedia berhenti menggunakan produk segera. Efek samping dapat dilaporkan ke FDA melalui program pelaporan efek samping MedWatch -nya.

Fasilitas Biosimilars Baru

Dalam berita Sandoz lainnya, perusahaan tersebut memecah fasilitas baru di Slovenia pada bulan Juli yang akan menghasilkan produk suntik untuk biosimilar yang ada dan yang akan datang. Proyek ini menambahkan $ 440 juta untuk investasi $ 1,1 miliar yang telah direncanakan perusahaan hingga 2029.

“Biosimilar adalah segmen paling cepat dari pipa kami karena kebutuhan pasien dan sistem perawatan kesehatan untuk obat-obatan kritis ini terus tumbuh dengan cepat,” Richard Saynor, kepala eksekutif Sandoz, mengatakan dalam siaran pers (2). “Sebagai pemimpin global di lapangan, kami berinvestasi untuk memenuhi permintaan pasien yang berkembang pesat. Dengan komitmen lebih dari US $ 1,1 miliar, kami bangga memperluas kapasitas manufaktur biosimilar kami di Eropa sebagai investor asing langsung Slovenia. Ini adalah langkah besar lainnya yang akan memposisikan Sandoz yang unik untuk mengkapitalisasi biosimilaries yang belum pernah ada di depan Sandoz.

“Investasi kami dalam bidang bioman di jantung Eropa menandai tonggak penting lainnya dalam membangun jaringan manufaktur independen kami sendiri – yang meningkatkan kontrol, ketahanan, dan kelincahan kami di seluruh rantai pasokan global,” Glenn Gerecke, kepala manufaktur dan petugas pasokan di Sandoz, mengatakan dalam rilisnya. “Kami mengakui nilai strategis yang dibawa Slovenia bagi kami sebagai perusahaan: kumpulan bakat yang sangat terampil dalam ilmu pengetahuan alam dan teknik; lokasi sentral dengan infrastruktur logistik kelas dunia; produksi kompetitif biaya; dan lingkungan yang stabil dan berbasis inovasi yang terkait erat dengan akademia dan penelitian.”

Referensi

  1. FDA. Sandoz Inc. mengeluarkan penarikan kembali secara nasional secara nasional dari satu lot cefazolin untuk suntikan karena kesalahan pengemasan produk. Siaran pers. 27 Juni 2025. Https://www.fda.gov/safety/recalls-market-withdrawals-safety-alerts/sandoz-inc-issues-voluntary-nationwide-recall-oe-lot-cefazolin-injection-product-pendamping
  2. Sandoz. Sandoz lebih lanjut menegaskan kepemimpinan dalam biosimilars, memecah dasar fasilitas Slovenia baru untuk memperluas hub biosimilar Eropa dan jangkauan pasar global. Siaran pers. 1 Juli 2025. Https://www.sandoz.com/sandoz-further-asserts-leadership-biosimilars-break-ground-new-lovenia-facility-expand-uropean/